1
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2025; 62:1631-1674. [PMID: 39012443 PMCID: PMC11772559 DOI: 10.1007/s12035-024-04333-y] [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: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
2
|
Tavares ÉAF, de Souza DLS, da Silva Gomes FT, Holanda MVF, Oliveira RF, Paiva KM, de Gois Morais PLA, de Vasconcelos CL, de Paiva Cavalcanti DML, de Paiva Cavalcanti JRL. Calcium-Binding Proteins in the Autistic Brain-Potential Links to Symptom Development. Int J Dev Neurosci 2025; 85:e10412. [PMID: 39777736 DOI: 10.1002/jdn.10412] [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: 09/18/2024] [Revised: 12/13/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
Autism spectrum disorder (ASD) is a complex challenge, influenced by genetic and environmental factors. This review focuses on the proteins calbindin (CB), calretinin (CR) and parvalbumin (PV) in the context of ASD, exploring their clinical correlations and providing a deeper understanding of the spectrum. In addition, we seek to understand the role of these proteins in GABAergic regulation and their implication in the pathophysiology of ASD. The studies reviewed revealed a significant decrease in PV+ interneurons in the prefrontal cortex of individuals with ASD compared with typical controls. This reduction is associated with local synaptic hyperconnectivity, characterized by an increase in the number of excitatory synapses and a reduction in inhibitory synapses. A correlation was also observed between the decrease in PV+ and the severity of the behavioural symptoms of ASD. The research highlights GABAergic imbalance as a crucial component in the neuropathology of ASD, highlighting the role of calcium-binding proteins, especially PV, in regulating neuronal excitability and modulating synaptic connectivity. These findings provide valuable insights for the development of new therapeutic strategies intended to modulate neuronal activity and improve the symptoms associated with ASD.
Collapse
Affiliation(s)
- Élyssa Adriolly Freitas Tavares
- Faculty of Health Sciences, Department of Biomedical Sciences, State University of Rio Grande do Norte, Mossoró, Rio Grande do Norte, Brazil
| | - Débora Lopes Silva de Souza
- Faculty of Health Sciences, Department of Biomedical Sciences, State University of Rio Grande do Norte, Mossoró, Rio Grande do Norte, Brazil
| | - Francisca Tayná da Silva Gomes
- Faculty of Health Sciences, Department of Biomedical Sciences, State University of Rio Grande do Norte, Mossoró, Rio Grande do Norte, Brazil
| | - Maria Vanessa Freitas Holanda
- Faculty of Health Sciences, Department of Biomedical Sciences, State University of Rio Grande do Norte, Mossoró, Rio Grande do Norte, Brazil
| | - Rodrigo Freire Oliveira
- Faculty of Health Sciences, Department of Biomedical Sciences, State University of Rio Grande do Norte, Mossoró, Rio Grande do Norte, Brazil
| | - Karina Maia Paiva
- Faculty of Health Sciences, Department of Biomedical Sciences, State University of Rio Grande do Norte, Mossoró, Rio Grande do Norte, Brazil
| | | | - Cláudio Lopes de Vasconcelos
- Faculty of Exact and Natural Sciences, Department of Chemistry, State University of Rio Grande do Norte, Mossoró, Rio Grande do Norte, Brazil
| | - Diogo Manuel Lopes de Paiva Cavalcanti
- Center for Biomedical and Health Sciences, Department of Health Sciences, Federal Rural University of the Semi-Arid, Mossoró, Rio Grande do Norte, Brazil
| | | |
Collapse
|
3
|
Perovic M, Pavlovic D, Palmer Z, Udo MSB, Citadin CT, Rodgers KM, Wu CYC, Zhang Q, Lin HW, Tesic V. Modulation of GABAergic system as a therapeutic option in stroke. Exp Neurol 2025; 384:115050. [PMID: 39522803 DOI: 10.1016/j.expneurol.2024.115050] [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: 07/31/2024] [Revised: 10/18/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Stroke is one of the leading causes of death and permanent adult disability worldwide. Despite the improvements in reducing the rate and mortality, the societal burden and costs of treatment associated with stroke management are increasing. Most of the therapeutic approaches directly targeting ischemic injury have failed to reduce short- and long-term morbidity and mortality and more effective therapeutic strategies are still needed to promote post-stroke functional recovery. Decades of stroke research have been focused on hyperexcitability and glutamate-induced excitotoxicity in the acute phase of ischemia and their relation to motor deficits. Recent advances in understanding the pathophysiology of stroke have been made with several lines of evidence suggesting that changes in the neurotransmission of the major inhibitory system via γ-Aminobutyric acid (GABA) play a particularly important role in functional recovery and deserve further attention. The present review provides an overview of how GABAergic neurotransmission changes correlate with stroke recovery and outlines GABAergic system modulators with special emphasis on neurosteroids that have been shown to affect stroke pathogenesis or plasticity or to protect against cognitive decline. Supporting evidence from both animal and human clinical studies is presented and the potential for GABA signaling-targeted therapies for stroke is discussed to translate this concept to human neural repair therapies. Age and sex are considered crucial parameters related to the pathophysiology of stroke and important factors in the development of therapeutic pharmacological strategies. Future work is needed to deepen our knowledge of the neurochemical changes after stroke, extend the conceptual framework, and allow for the development of more effective interventions that include the modulation of the inhibitory system.
Collapse
Affiliation(s)
- Milka Perovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Damjan Pavlovic
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Zoe Palmer
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Mariana S B Udo
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Cristiane T Citadin
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Krista M Rodgers
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Celeste Yin-Chien Wu
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Vesna Tesic
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA.
| |
Collapse
|
4
|
Sakthivel K, Balasubramanian R, Sampathrajan V, Veerasamy R, Appachi SV, K K K. Transforming tomatoes into GABA-rich functional foods through genome editing: A modern biotechnological approach. Funct Integr Genomics 2025; 25:27. [PMID: 39871009 DOI: 10.1007/s10142-025-01538-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/29/2025]
Abstract
Gamma-aminobutyric acid (GABA) functions as an inhibitory neurotransmitter which blocks the impulses between nerve cells in the brain. Due to the increasing awareness about the health promoting benefits associated with GABA, it is also artificially synthesized and consumed as a nutritional supplement by people in some regions of the world. Though among the fresh vegetables, tomato fruits do contain a comparatively higher amount of GABA (0.07 to 2.01 mg g-1 FW), it needs to be further enhanced to fully impart its potential health benefits. Achieving this feat through classical breeding approaches is time and resource consuming, and is also associated with linkage drag. On the other hand, precise targeting of specific sites in the genome with less off- target effects is mediated by CRISPR/Cas9 genome editing tool and is widely used to overcome the barriers associated with traditional breeding approaches. Combining genome editing with speed breeding techniques can enable the rapid development of GABA-rich tomato cultivars, paving a way to unlock a new era of functional foods, where every bite contributes to cognitive well-being and holistic health. This review highlights the significance of GABA boosted functional foods and explores the potential of CRISPR/Cas9 technology for developing GABA enriched tomatoes.
Collapse
Affiliation(s)
- Kausalya Sakthivel
- Department of Plant Biotechnology, Tamilnadu Agricultural University, 641003, Coimbatore, India
| | | | | | - Ravichandran Veerasamy
- Department of Crop Physiology, Tamilnadu Agricultural University, 641003, Coimbatore, India
| | | | - Kumar K K
- Department of Plant Biotechnology, Tamilnadu Agricultural University, 641003, Coimbatore, India.
| |
Collapse
|
5
|
Zhou J, Noviello CM, Teng J, Moore H, Lega B, Hibbs RE. Resolving native GABA A receptor structures from the human brain. Nature 2025:10.1038/s41586-024-08454-1. [PMID: 39843743 DOI: 10.1038/s41586-024-08454-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/26/2024] [Indexed: 01/24/2025]
Abstract
Type A GABA (γ-aminobutyric acid) receptors (GABAA receptors) mediate most fast inhibitory signalling in the brain and are targets for drugs that treat epilepsy, anxiety, depression and insomnia and for anaesthetics1,2. These receptors comprise a complex array of 19 related subunits, which form pentameric ligand-gated ion channels. The composition and structure of native GABAA receptors in the human brain have been inferred from subunit localization in tissue1,3, functional measurements and structural analysis from recombinant expression4-7 and in mice8. However, the arrangements of subunits that co-assemble physiologically in native human GABAA receptors remain unknown. Here we isolated α1 subunit-containing GABAA receptors from human patients with epilepsy. Using cryo-electron microscopy, we defined a set of 12 native subunit assemblies and their 3D structures. We address inconsistencies between previous native and recombinant approaches, and reveal details of previously undefined subunit interfaces. Drug-like densities in a subset of these interfaces led us to uncover unexpected activity on the GABAA receptor of antiepileptic drugs and resulted in localization of one of these drugs to the benzodiazepine-binding site. Proteomics and further structural analysis suggest interactions with the auxiliary subunits neuroligin 2 and GARLH4, which localize and modulate GABAA receptors at inhibitory synapses. This work provides a structural foundation for understanding GABAA receptor signalling and targeted pharmacology in the human brain.
Collapse
Affiliation(s)
- Jia Zhou
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Colleen M Noviello
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Jinfeng Teng
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Haley Moore
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bradley Lega
- Departments of Neurological Surgery, Neurology, Psychiatry and the Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ryan E Hibbs
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Xu Y, Mortensen M, Liebowitz S, Jensen NN, Tian Y, Bavo F, Seidel T, Smart TG, Frølund B. Design, Synthesis, and Pharmacological Evaluation of Nonsteroidal Tricyclic Ligands as Modulators of GABA A Receptors. J Med Chem 2025. [PMID: 39808706 DOI: 10.1021/acs.jmedchem.4c02881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
GABAA receptors (GABAARs) are the major elements of inhibitory neurotransmission in the central nervous system (CNS). They are established targets for regulation by endogenous brain neuroactive steroids (NASs) such as pregnanolone. However, the complexity of de novo synthesis of NAS derivatives has hindered attempts to circumvent the principal limitations of using endogenous NASs, including selectivity and limited oral bioavailability. In this study, we designed a series of tricyclic compounds, inspired by the structures of pregnanolone and pregnenolone sulfate, to explore novel nonsteroidal alternatives. Using patch clamp electrophysiology, we demonstrate that these compounds exhibit either positive or negative allosteric modulation of GABAARs. Specifically, we discover a positive allosteric modulator (PAM) and a series of tricyclic sulfate-based negative allosteric modulators (NAMs) all active at the micromolar level. This research has significantly broadened the chemical diversity of ligands targeting GABAARs offering potential for efficacious allosteric modulators while avoiding the complexity of NAS synthesis.
Collapse
Affiliation(s)
- Yue Xu
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Martin Mortensen
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, U.K
| | - Seth Liebowitz
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, U.K
| | - Nicoline N Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Yongsong Tian
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Francesco Bavo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Thomas Seidel
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna 1090, Austria
| | - Trevor G Smart
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, U.K
| | - Bente Frølund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| |
Collapse
|
7
|
Vacher CM, Tsompanidis A, Firestein MR, Penn AA. Neuroactive steroid exposure impacts neurodevelopment: Comparison of human and rodent placental contribution. J Neuroendocrinol 2025:e13489. [PMID: 39789736 DOI: 10.1111/jne.13489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
The placenta is a fetal endocrine organ that secretes many neuroactive factors, including steroids, that play critical roles in brain development. The study of the placenta-brain axis and the links between placental function and brain development represents an emerging research area dubbed "neuroplacentology." The placenta drives many circulating fetal steroids to very high levels during gestation. Recent studies have highlighted the critical role of placental steroids in shaping specific brain structures and behaviors. This review uses a cross-species framework to discuss the genomic factors, in-utero environmental changes, and placental conditions that alter placental steroidogenesis, leading to changes in early developmental trajectories relevant for psychiatric conditions such as autism, in a sex-linked manner.
Collapse
Affiliation(s)
- Claire-Marie Vacher
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Morgan R Firestein
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, USA
| | - Anna A Penn
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
- New York Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| |
Collapse
|
8
|
Toraldo DM, Palma Modoni A, Scoditti E, De Nuccio F. Obstructive sleep apnoea as a neuromuscular respiratory disease arising from an excess of central GABAergic neurotransmitters: a new disease model. Front Cell Neurosci 2025; 18:1429570. [PMID: 39835289 PMCID: PMC11743696 DOI: 10.3389/fncel.2024.1429570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Affiliation(s)
- Domenico Maurizio Toraldo
- Respiratory Care Unit, Rehabilitation Department, “V. Fazzi” Hospital, Azienda Sanitaria Locale, San Cesario, Lecce, Italy
| | - Alessandra Palma Modoni
- Respiratory Care Unit, Rehabilitation Department, “V. Fazzi” Hospital, Azienda Sanitaria Locale, San Cesario, Lecce, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Institute of Clinical Physiology (IFC), Lecce, Italy
| | - Francesco De Nuccio
- Laboratory of Human Anatomy, Department of Experimental Medicine, University of the Salento, Lecce, Italy
| |
Collapse
|
9
|
Wang Y, Zhang Y, Li W, Salovska B, Zhang J, Li T, Li H, Liu Y, Kaczmarek LK, Pusztai L, Klein DE. GABA A receptor π forms channels that stimulate ERK through a G-protein-dependent pathway. Mol Cell 2025; 85:166-176.e5. [PMID: 39642883 PMCID: PMC11698630 DOI: 10.1016/j.molcel.2024.11.016] [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: 02/08/2024] [Revised: 05/03/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024]
Abstract
The rare γ-aminobutyric acid type-A receptor (GABAAR) subunit π (GABRP) is highly expressed in certain cancers, where it stimulates growth through extracellular-regulated kinase (ERK) signaling by an uncharacterized pathway. To elucidate GABRP's signaling mechanism, we determined cryoelectron microscopy (cryo-EM) structures of GABRP embedded in native nanodiscs, both in the presence and absence of GABA. Structurally, GABRP homopentamers closely resemble heteropentameric GABAAR anion channels, transitioning from a closed "resting" state to an open "active" state upon GABA binding. However, functional assays reveal that GABRP responds more like a type-B metabotropic receptor. At physiological concentrations of GABA, chloride flux is not detected. Rather, GABRP activates a G-protein-coupled pathway leading to ERK signaling. Ionotropic activity is only triggered at supraphysiological GABA concentrations, effectively decoupling it from GABRP's signaling functions. These findings provide a structural and functional blueprint for GABRP, opening new avenues for targeted inhibition of GABA growth signals in GABRP-positive cancers.
Collapse
Affiliation(s)
- Yueyue Wang
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Breast Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yalan Zhang
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Wenxue Li
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Barbora Salovska
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jianan Zhang
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tongqing Li
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Hengyi Li
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yansheng Liu
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Biomedical Informatics & Data Science, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lajos Pusztai
- Breast Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Daryl E Klein
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
10
|
Fujioka Y, Shiura H, Ishii M, Ono R, Endo T, Kiyonari H, Hirate Y, Ito H, Kanai-Azuma M, Kohda T, Kaneko-Ishino T, Ishino F. Targeting of retrovirus-derived Rtl8a/ 8b causes late-onset obesity, reduced social response and increased apathy-like behaviour. Open Biol 2025; 15:240279. [PMID: 39875098 PMCID: PMC11774587 DOI: 10.1098/rsob.240279] [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: 09/26/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 01/30/2025] Open
Abstract
Retrotransposon Gag-like (RTL) 8A, 8B and 8C are eutherian-specific genes derived from a certain retrovirus. They cluster as a triplet of genes on the X chromosome, but their function remains unknown. Here, we demonstrate that Rtl8a and Rtl8b play important roles in the brain: their double knockout (DKO) mice not only exhibit reduced social responses and increased apathy-like behaviour, but also become obese from young adulthood, similar to patients with late Prader-Willi syndrome (PWS), a neurodevelopmental genomic imprinting disorder. Mouse RTL8A/8B proteins are expressed in the prefrontal cortex and hypothalamus and localize to both the nucleus and cytoplasm of neurons, presumably due to the N-terminal nuclear localization signal-like sequence at the N-terminus. An RNAseq study in the cerebral cortex revealed reduced expression of several GABA type A receptor subunit genes in DKO, in particular Gabrb2, which encodes its β2 subunit. We confirmed the reduction of GABRB2 protein in the DKO cerebral cortex by western blotting. As GABRB2 has been implicated in the aetiology of several neurodevelopmental and neuropsychiatric disorders, it is likely that the reduction of GABRB2 is one of the major causes of the neuropsychiatric defects in the DKO mice.
Collapse
Affiliation(s)
- Yoshifumi Fujioka
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| | - Hirosuke Shiura
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Faculty of Life and Environmental Sciences, University of Yamanashi, Kohfu,Yamanashi 400-8510, Japan
| | - Masayuki Ishii
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
| | - Ryuichi Ono
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences (NIHS), Kawasaki, Kanagawa210-9501, Japan
| | - Tsutomu Endo
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo650-0047, Japan
| | | | - Hikaru Ito
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
- Research Facility Center for Science and Technology, Kagawa University, Takamatsu,Kagawa 761-0793, Japan
| | | | - Takashi Kohda
- Faculty of Life and Environmental Sciences, University of Yamanashi, Kohfu,Yamanashi 400-8510, Japan
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, Tokai University School of Medicine, Isehara, Kanagawa259-1193, Japan
| | - Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| |
Collapse
|
11
|
Bukanova JV, Kondratenko RV, Solntseva EI. Interaction Between Allopregnanolone and Amiloride Binding Sites on the GABA A Receptor. Cell Biochem Biophys 2024:10.1007/s12013-024-01654-6. [PMID: 39730891 DOI: 10.1007/s12013-024-01654-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
Allopregnanolone (Allo) is a positive allosteric modulator of the GABAA receptor, and amiloride (Ami) is a competitive antagonist of the GABAA receptor. The purpose of this work was to study the combined effect of Allo and Ami on functional activity of GABAA receptor. The GABA-induced chloride current (IGABA) was measured in isolated Purkinje cells of rat cerebellum using the patch-clamp technique and a system of fast application. Our results indicate that Allo suppresses the inhibitory effect of Ami on IGABA, the IC50 value of Ami concentration-response curve was increased from 164 to 547 µM (P < 0.001) in the presence of Allo. Next, GABA concentration-response curves (EC50 = 5.8 µM) were constructed in the presence of Allo (EC50 = 1.2 µM), Ami (EC50 = 25.5 µM), and the combination of Allo+Ami (EC50 = 3.2 µM). Changes in EC50 values as a percentage relative to the control were calculated. The blocking effect of Ami is reduced in the presence of Allo (340% vs 150%, P < 0.01) and the potentiating effect of Allo does not change in the presence of Ami (78% vs 87%, P > 0.05). The results suggest that there is an allosteric relationship between the Allo and Ami binding sites on GABAA receptor that operates in one direction, from Allo sites to Ami site, but not vice versa.
Collapse
|
12
|
Singh M, Indurthi DC, Mittal L, Auerbach A, Asthana S. Conformational dynamics of a nicotinic receptor neurotransmitter site. eLife 2024; 13:RP92418. [PMID: 39693137 DOI: 10.7554/elife.92418] [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] [Indexed: 12/19/2024] Open
Abstract
Agonists enhance receptor activity by providing net-favorable binding energy to active over resting conformations, with efficiency (η) linking binding energy to gating. Previously, we showed that in nicotinic receptors, η-values are grouped into five structural pairs, correlating efficacy and affinity within each class, uniting binding with allosteric activation (Indurthi and Auerbach, 2023). Here, we use molecular dynamics (MD) simulations to investigate the low-to-high affinity transition (L→H) at the Torpedo α-δ nicotinic acetylcholine receptor neurotransmitter site. Using four agonists spanning three η-classes, the simulations reveal the structural basis of the L→H transition where: the agonist pivots around its cationic center ('flip'), loop C undergoes staged downward displacement ('flop'), and a compact, stable high-affinity pocket forms ('fix'). The η derived from binding energies calculated in silico matched exact values measured experimentally in vitro. Intermediate states of the orthosteric site during receptor activation are apparent only in simulations, but could potentially be observed experimentally via time-resolved structural studies.
Collapse
Affiliation(s)
- Mrityunjay Singh
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center,Translational Health Science and Technology Institute, Faridabad, India
| | - Dinesh C Indurthi
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, Buffalo, United States
| | - Lovika Mittal
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center,Translational Health Science and Technology Institute, Faridabad, India
| | - Anthony Auerbach
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, Buffalo, United States
| | - Shailendra Asthana
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center,Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
13
|
Zhang Q, Zhu L, Li H, Chen Q, Li N, Li J, Zhao Z, Xiao D, Tang T, Bi C, Zhang Y, Zhang H, Zhang G, Li M, Zhu Y, Zhang J, Kong J. Insights and progress on the biosynthesis, metabolism, and physiological functions of gamma-aminobutyric acid (GABA): a review. PeerJ 2024; 12:e18712. [PMID: 39703920 PMCID: PMC11657192 DOI: 10.7717/peerj.18712] [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: 08/19/2024] [Accepted: 11/24/2024] [Indexed: 12/21/2024] Open
Abstract
GABA (γ-aminobutyric acid) is a non-protein amino acid that occurs naturally in the human brain, animals, plants and microorganisms. It is primarily produced by the irreversible action of glutamic acid decarboxylase (GAD) on the α-decarboxylation of L-glutamic acid. As a major neurotransmitter in the brain, GABA plays a crucial role in behavior, cognition, and the body's stress response. GABA is mainly synthesized through the GABA shunt and the polyamine degradation pathways. It works through three receptors (GABAA, GABAB, and GABAC), each exhibiting different pharmacological and physiological characteristics. GABA has a variety of physiological roles and applications. In plants, it regulates growth, development and stress responses. In mammals, it influences physiological functions such as nervous system regulation, blood pressure equilibrium, liver and kidneys enhancement, hormone secretion regulation, immunity enhancement, cancer prevention, as well as anti-aging effects. As a biologically active ingredient, GABA possesses unique physiological effects and medicinal value, leading to its widespread application and substantially increased market demand in the food and pharmaceutical industries. GABA is primarily produced through chemical synthesis, plant enrichment and microbial fermentation. In this review, we first make an overview of GABA, focusing on its synthesis, metabolism, GABA receptors and physiological functions. Next, we describe the industrial production methods of GABA. Finally, we discuss the development of ligands for the GABA receptor binding site, the prospects of GABA production and application, as well as its clinical trials in potential drugs or compounds targeting GABA for the treatment of epilepsy. The purpose of this review is to attract researchers from various fields to focus on GABA research, promote multidisciplinary communications and collaborations, break down disciplinary barriers, stimulate innovative research ideas and methods, and advance the development and application of GABA in medicine, agriculture, food and other fields.
Collapse
Affiliation(s)
- Qingli Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Lei Zhu
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Hailong Li
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Qu Chen
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Nan Li
- Department of Rehabilitation, Qingdao Binhai College Affiliated Hospital, Qingdao, China
| | - Jiansheng Li
- Department of Nephrology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Zichu Zhao
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Di Xiao
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Tingting Tang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Chunhua Bi
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Yan Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Haili Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Guizhen Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Mingyang Li
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Yanli Zhu
- College of Life Sciences and Engineering, Northwest Minzu University, Lanzhou, China
| | - Jingjing Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Jingjing Kong
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| |
Collapse
|
14
|
Tian L, Li XH, Zhao YL, Yi HY, Liu XR, Yao R, Hou XM, Zhu X, Huo FQ, Chen T, Liang L. DNMT3a Downregulation Ttriggered Upregulation of GABA A Receptor in the mPFC Promotes Paclitaxel-Induced Pain and Anxiety in Male Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407387. [PMID: 39679872 DOI: 10.1002/advs.202407387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Chemotherapeutic agents, such as paclitaxel (PTX), induce neuroplastic changes and alter gene expression in the prefrontal cortex (PFC), which may be associated with chemotherapy-induced pain and negative emotions. Notably, DNA methylation undergoes adaptive changes in neurological disorders, emerging as a promising target for neuromodulation. In this study, systemic administration of PTX leads to a decrease in the expression of the DNA methyltransferase DNMT3a, while concurrently upregulating the expression of Gabrb1 mRNA and its encoded GABAARβ1 protein in the medial PFC (mPFC) of male mice. Overexpression of DNMT3a in the mPFC alleviates PTX-induced pain hypersensitivity, and anxiety-like behavior in these mice. Additionally, it reverses the PTX-induced increase in inhibitory synaptic transmission in the pyramidal neurons of the mPFC. Mechanistically, the upregulation of GABAARβ1 in the mPFC is linked to the reduced expression of DNMT3a and DNA hypomethylation at the promoter region of the Gabrb1 gene. Furthermore, a long-term diet rich in methyl donors alleviates PTX-induced pain hypersensitivity and anxiety-like behavior in mice. These findings suggest that the DNMT3a-mediated upregulation of GABAARβ1 in the mPFC contributes to PTX-induced neuropathic pain and anxiety, highlighting DNA methylation-dependent epigenetic regulation as a potential therapeutic target for addressing chemotherapy-induced cortical dysfunction.
Collapse
Affiliation(s)
- Lixia Tian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Xu-Hui Li
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China
| | - Yu-Long Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Hui-Yuan Yi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Xue-Ru Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Rongrong Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Xue-Mei Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Xuan Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Fu-Quan Huo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Tao Chen
- Department of Anatomy and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Lingli Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| |
Collapse
|
15
|
Long H, Wu H, Sun C, Xu X, Yang XH, Xiao J, Lv M, Chen Q, Fan M. Biological mechanism of sex differences in mental rotation: Evidence from multimodal MRI, transcriptomic and receptor/transporter data. Neuroimage 2024; 304:120955. [PMID: 39586343 DOI: 10.1016/j.neuroimage.2024.120955] [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: 08/09/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024] Open
Abstract
Sex differences in mental rotation are a well-documented phenomenon in cognitive research, with implications for the differing prevalence of neuropsychiatric disorders such as autism spectrum disorder (ASD), Alzheimer's disease (AD) and major depressive disorder (MDD) between the sexes. Despite extensive documentation, the biological mechanism underpinning these differences remain elusive. This study aimed to elucidate neural, genetic, and molecular bases of these disparities in mental rotation by integrating data from multimodal magnetic resonance imaging (MRI), transcriptomic and receptor/transporter. We first calculated the dynamic regional homogeneity (dReHo), gray matter volume (GMV) and fractional anisotropy (FA) in voxel-wise manner and parceled them into 246 brain regions based on Brainnetome Atlas. Subsequent analyses involved Pearson Correlations to examine the association between mental rotation performance and dReHo/GMV/FA and two-sample t-tests to delineate gender differences in these indices. Based on the above results, further mediation analysis was conducted to explore the relationship between sex, brain biomarkers and mental rotation. In addition, transcriptome-neuroimaging association analysis and correlation analysis between brain biomarkers and neurotransmitter receptor/transporter distribution were also performed to uncover genetic and molecular mechanisms contributing to the observed sex differences in mental rotation. We found correlations between mental rotation performance and dReHo, GMV and FA of the inferior parietal lobule (IPL) and superior temporal gyrus (STG) and sex effects on these brain biomarkers. Notably, the dReHo of the left IPL mediated the relationship between sex and mental rotation. Further correlation analysis revealed that the proton-coupled oligopeptide transporter PEPT2 (SLC15A2) and interleukin 17 receptor D (IL17RD) were associated with sex-related t-statistic maps and mental rotation-related r-statistic maps of dReHo. Moreover, γ-aminobutyric acid subtype A (GABAA) receptor availability was correlated with the r-statistic of dReHo, while norepinephrine transporter (NET) availability was correlated with its t-statistic. Serial mediation models revealed the indirect effect of these genes on the r-statistic maps through the transporter/receptor and t-statistic maps. Our findings provide novel insights into the biological mechanism underlying sex differences in mental rotation, identifying potential biomarkers for cognitive impairment and explaining variations in prevalence of certain mental disorders between the sexes. These results highlight the necessity of considering sex in research on mental health disorders.
Collapse
Affiliation(s)
- Haixia Long
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Hao Wu
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Chaoliang Sun
- Zhejiang Lab, Zhongtai Street, Yuhang District, Hangzhou 311100, China
| | - Xinli Xu
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Xu-Hua Yang
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Jie Xiao
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Mingqi Lv
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, China
| | - Qiuju Chen
- School of Design and Architecture, Zhejiang University of Technology, Hangzhou 310023, China
| | - Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, China.
| |
Collapse
|
16
|
Maloum-Rami F, Cheung P, Antoni G, Jin Z, Eriksson O, Espes D. PET imaging of GABA A receptors in pancreatic islets by [ 11C]flumazenil. EJNMMI Res 2024; 14:122. [PMID: 39623212 PMCID: PMC11612099 DOI: 10.1186/s13550-024-01176-5] [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: 08/27/2024] [Accepted: 11/05/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease characterized by a progressive β-cell destruction. There are no clinically established methods for quantifying endocrine cells of the pancreas and current knowledge is almost exclusively based on autopsy material and functional measurements. Based on the expression of the γ-aminobutyric acid A receptors (GABAARs) in pancreatic islets and the fact that GABAAR agonists are being explored as treatment for T1D, we hypothesized that the positron emission tomography (PET) tracer [11C]flumazenil ([11C]FMZ) could serve as a marker of the endocrine mass of the pancreas. The in vivo uptake of [11C]FMZ in pig pancreas was evaluated by PET/CT, either tracer alone or after blockade of GABAAR by unlabeled flumazenil. The pancreatic binding of [11C]FMZ was investigated in vitro with frozen sections of pig pancreas as well as human organ donors, in addition to isolated mouse and human islets and exocrine preparations. The expression of GABAAR subunits in pig, human and mouse pancreas was explored by immunohistochemistry. RESULTS Strong specific in vivo uptake of [11C]FMZ was observed in the pig brain as expected, but in the pancreas the signal was moderate and only partially reduced by blockade. In vitro experiments revealed a positive but weak and variable binding of [11C]FMZ in islets compared to exocrine tissue in the mouse, pig and human pancreas. In pig and mouse pancreatic islets we identified the GABAAR subunits β2 and γ2 but not α2. In the human pancreas from non-diabetic donors, we have identified the α2, β2 (although weak) and γ2 subunits, whereas from a T2D donor the α2 subunit was missing. CONCLUSIONS Our findings suggest that [11C]FMZ bind to GABAARs in the islets, but not with a sufficient contrast or magnitude to be implemented as an in vivo PET marker for the endocrine mass of the pancreas. However, GABAARs with different subunits are widely expressed in the endocrine cells within the pancreas in pig, human and mouse. Hence, the GABAAR could still be a potential imaging target for the endocrine cells of the pancreas but would require tracers with higher affinity and selectivity for individual GABAAR subunits.
Collapse
Affiliation(s)
- Faïza Maloum-Rami
- Department of Medical Cell Biology, Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Box 571, 75123, Uppsala, Sweden
| | - Pierre Cheung
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Dag Hammarskjölds Väg 14C, 3Tr, 75183, Uppsala, Sweden
| | - Gunnar Antoni
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Dag Hammarskjölds Väg 14C, 3Tr, 75183, Uppsala, Sweden
- PET Center, Center for Medical Imaging, Uppsala University Hospital, Uppsala, Sweden
| | - Zhe Jin
- Department of Medical Cell Biology, Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Box 571, 75123, Uppsala, Sweden
| | - Olof Eriksson
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Dag Hammarskjölds Väg 14C, 3Tr, 75183, Uppsala, Sweden.
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| | - Daniel Espes
- Department of Medical Cell Biology, Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Box 571, 75123, Uppsala, Sweden.
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
17
|
Klett N, Gompf HS, Allen CN, Cravetchi O, Hablitz LM, Gunesch AN, Irwin RP, Todd WD, Saper CB, Fuller PM. GABAergic signalling in the suprachiasmatic nucleus is required for coherent circadian rhythmicity. Eur J Neurosci 2024; 60:6652-6667. [PMID: 39558544 PMCID: PMC11612841 DOI: 10.1111/ejn.16582] [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: 02/20/2024] [Revised: 09/11/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024]
Abstract
The suprachiasmatic nucleus is the circadian pacemaker of the mammalian brain. Suprachiasmatic nucleus neurons display synchronization of their firing frequency on a circadian timescale, which is required for the pacemaker function of the suprachiasmatic nucleus. However, the mechanisms by which suprachiasmatic nucleus neurons remain synchronized in vivo are poorly understood, although synaptic communication is considered indispensable. Suprachiasmatic nucleus neurons contain the neurotransmitter GABA and express GABA receptors. This has inspired the hypothesis that GABA signalling may play a central role in network synchronization, although this remains untested in vivo. Here, using local genetic deletion, we show that disruption of GABA synaptic transmission within the suprachiasmatic nucleus of adult mice results in the eventual deterioration of physiological and behavioural rhythmicity in vivo and concomitant cellular desynchrony in vitro. These findings suggest that intercellular GABA signalling is essential for behavioural rhythmicity and cellular synchrony of the suprachiasmatic nucleus neural network.
Collapse
Affiliation(s)
- Nathan Klett
- Oregon Institute for Occupational Health SciencesUSA
- Neuroscience Graduate ProgramUSA
| | - Heinrich S. Gompf
- Department of Neurological SurgeryUniversity of California, DavisDavisCAUSA
| | - Charles N. Allen
- Oregon Institute for Occupational Health SciencesUSA
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
| | | | - Lauren M. Hablitz
- Oregon Institute for Occupational Health SciencesUSA
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
- Present address:
Center for Translational NeuromedicineUniversity of Rochester Medical CenterRochesterNYUSA
| | | | | | - William D. Todd
- Department of Neurology, Division of Sleep Medicine, and Program in NeuroscienceBeth Israel Deaconess Medical Center, Harvard Medical School BostonMAUSA
- Present address:
Department of Zoology and PhysiologyUniversity of WyomingLaramieWYUSA
| | - Clifford B. Saper
- Department of Neurology, Division of Sleep Medicine, and Program in NeuroscienceBeth Israel Deaconess Medical Center, Harvard Medical School BostonMAUSA
| | - Patrick M. Fuller
- Department of Neurological SurgeryUniversity of California, DavisDavisCAUSA
| |
Collapse
|
18
|
Yu Z, Chen X, Liu Z, Ding R, Xu J. Subunit-specific mechanisms of isoflurane-induced acute tonic inhibition in dentate gyrus granule neuron. Exp Biol Med (Maywood) 2024; 249:10171. [PMID: 39529664 PMCID: PMC11550974 DOI: 10.3389/ebm.2024.10171] [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: 03/17/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Prolonged exposure to volatile anesthetics may raise the risk of developing cognitive impairment by acting on gamma-a Aminobutyric acid A receptors (GABAAR). The dentate gyrus plays an important role in the hippocampus and has a high potential for neural plasticity. However, it is unknown whether prolonged anesthesia induces a change in acute phasic or tonic inhibition in dentate gyrus granule cells (DGGCs) by acting on GABAAR. In order to verify the effects of volatile anesthetics on the current in DGGCs, a whole-cell patch was applied to record acute brain slices, and this study indicated that 4 h but not 2 h of isoflurane (ISO) exposure induced significantly larger tonic currents in DGGCs other than hippocampal CA1 pyramidal and thalamic relay neurons. Furthermore, this study demonstrated that the increased tonic current in DGGCs was dependent on the δ subunit-containing GABAARs by using transgenic δ subunit knockout mice. In conclusion, the δ subunit specific GABAAR is the key element that increased acute tonic inhibition in DGGCs of mice after prolonged ISO exposure, which may be one of the mechanisms of ISO neurotoxicity to the developing brain.
Collapse
Affiliation(s)
- Zhiqiang Yu
- Department of Anesthesiology, Tianjin Central Hospital for Gynecology and Obstetrics, Tianjin, China
| | - Xiaodan Chen
- Department of Anesthesiology, Tianjin Central Hospital for Gynecology and Obstetrics, Tianjin, China
| | - Zheng Liu
- Department of Anesthesiology, Tianjin Central Hospital for Gynecology and Obstetrics, Tianjin, China
| | - Ran Ding
- Tianjin Research Institute of Anesthesiology and Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jin Xu
- Department of Anesthesiology, Tianjin Hospital, Tianjin, China
| |
Collapse
|
19
|
Zhang W, Liu T, Li J, Singh J, Chan A, Islam A, Petrache A, Peng Y, Harvey K, Ali AB. Decreased extrasynaptic δ-GABA A receptors in PNN-associated parvalbumin interneurons correlates with anxiety in APP and tau mouse models of Alzheimer's disease. Br J Pharmacol 2024; 181:3944-3975. [PMID: 38886118 DOI: 10.1111/bph.16441] [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: 03/15/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is associated with gradual memory loss and anxiety which affects ~75% of AD patients. This study investigated whether AD-associated anxiety correlated with modulation of extrasynaptic δ-subunit-containing GABAA receptors (δ-GABAARs) in experimental mouse models of AD. EXPERIMENTAL APPROACH We combined behavioural experimental paradigms to measure cognition performance, and anxiety with neuroanatomy and molecular biology, using familial knock-in (KI) mouse models of AD that harbour β-amyloid (Aβ) precursor protein App (AppNL-F) with or without humanized microtubule-associated protein tau (MAPT), age-matched to wild-type control mice at three different age windows. RESULTS AppNL-F KI and AppNL-F/MAPT AD models showed a similar magnitude of cognitive decline and elevated magnitude of anxiety correlated with neuroinflammatory hallmarks, including triggering receptor expressed on myeloid cells 2 (TREM2), reactive astrocytes and activated microglia consistent with accumulation of Aβ, tau and down-regulation of Wnt/β-catenin signalling compared to aged-matched WT controls. In both the CA1 region of the hippocampus and dentate gyrus, there was an age-dependent decline in the expression of δ-GABAARs selectively expressed in parvalbumin (PV)-expressing interneurons, encapsulated by perineuronal nets (PNNs) in the AD mouse models compared to WT mice. In vivo positive allosteric modulation of the δ-GABAARs, using a δ-selective-compound DS2, decreased the level of anxiety in the AD mouse models, which was correlated with reduced hallmarks of neuroinflammation, and 'normalisation' of the expression of δ-GABAARs. CONCLUSIONS Our data show that the δ-GABAARs could potentially be targeted for alleviating symptoms of anxiety, which would greatly improve the quality of life of AD individuals.
Collapse
|
20
|
Lyu Q, Xue W, Liu R, Ma Q, Kasaragod VB, Sun S, Li Q, Chen Y, Yuan M, Yang Y, Zhang B, Nie A, Jia S, Shen C, Gao P, Rong W, Yu C, Bi Y, Zhang C, Nan F, Ning G, Rao Z, Yang X, Wang J, Wang W. A brain-to-gut signal controls intestinal fat absorption. Nature 2024; 634:936-943. [PMID: 39261733 DOI: 10.1038/s41586-024-07929-5] [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: 01/28/2022] [Accepted: 08/08/2024] [Indexed: 09/13/2024]
Abstract
Although fat is a crucial source of energy in diets, excessive intake leads to obesity. Fat absorption in the gut is prevailingly thought to occur organ-autonomously by diffusion1-3. Whether the process is controlled by the brain-to-gut axis, however, remains largely unknown. Here we demonstrate that the dorsal motor nucleus of vagus (DMV) plays a key part in this process. Inactivation of DMV neurons reduces intestinal fat absorption and consequently causes weight loss, whereas activation of the DMV increases fat absorption and weight gain. Notably, the inactivation of a subpopulation of DMV neurons that project to the jejunum shortens the length of microvilli, thereby reducing fat absorption. Moreover, we identify a natural compound, puerarin, that mimics the suppression of the DMV-vagus pathway, which in turn leads to reduced fat absorption. Photoaffinity chemical methods and cryogenic electron microscopy of the structure of a GABAA receptor-puerarin complex reveal that puerarin binds to an allosteric modulatory site. Notably, conditional Gabra1 knockout in the DMV largely abolishes puerarin-induced intestinal fat loss. In summary, we discover that suppression of the DMV-vagus-jejunum axis controls intestinal fat absorption by shortening the length of microvilli and illustrate the therapeutic potential of puerarin binding to GABRA1 in fat loss.
Collapse
Affiliation(s)
- Qianqian Lyu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, SJTUSM, Shanghai, China
| | - Wenzhi Xue
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Clinical Neuroscience Center, Ruijin Hospital Luwan Branch, SJTUSM, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, SJTUSM, Shanghai, China
| | - Qinyun Ma
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, SJTUSM, Shanghai, China
| | | | - Shan Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Qian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yanru Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Mingyang Yuan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yuying Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Bing Zhang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Aifang Nie
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, SJTUSM, Shanghai, China
| | - Sheng Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Chongrong Shen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Po Gao
- Department of Anatomy and Physiology, SJTUSM, Shanghai, China
| | - Weifang Rong
- Department of Anatomy and Physiology, SJTUSM, Shanghai, China
| | - Chenxi Yu
- The Chinese National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, SJTUSM, Shanghai, China
| | - Chunlei Zhang
- Institut Pasteur, Université de Paris, Neural Circuits for Spatial Navigation and Memory, Paris, France
| | - Fajun Nan
- The Chinese National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, SJTUSM, Shanghai, China
| | - Zihe Rao
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Xiuna Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, SJTUSM, Shanghai, China.
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, SJTUSM, Shanghai, China.
| |
Collapse
|
21
|
Valladares-Hernández IU, Hernández-Martínez JM, Cuaxospa JM, Jiménez-Vázquez EN, Sánchez-Jaramillo E, Arias JM, García U. Molecular cloning, functional characterization and differential expression of two novel GABA AR-like subunits from red swamp crayfish Procambarus clarkii. Eur J Neurosci 2024; 60:5980-5999. [PMID: 39275952 DOI: 10.1111/ejn.16540] [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: 07/19/2023] [Revised: 08/09/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024]
Abstract
In this work, we cloned and functionally expressed two novel GABAA receptor subunits from Procambarus clarkii crayfish. These two new subunits, PcGABAA-α and PcGABAA-β2, revealed significant sequence homology with the PcGABAA-β subunit, previously identified in our laboratory. In addition, PcGABAA-α subunit also shared a significant degree of identity with the Drosophila melanogaster genes DmGRD (GABA and glycine-like receptor subunits of Drosophila) as well as PcGABAA-β2 subunit with DmLCCH3 (ligand-gated chloride channel homolog 3). Electrophysiological recordings showed that the expression in HEK cells of the novel subunits, either alone or in combination, failed to form functional homo- or heteromeric receptors. However, the co-expression of PcGABAA-α with PcGABAA-β evoked sodium- or chloride-dependent currents that accurately reproduced the time course of the GABA-evoked currents in the X-organ neurons from crayfish, suggesting that these GABA subunits combine to form two types of GABA receptors, one with cationic selectivity filter and the other preferentially permeates anions. On the other hand, PcGABAA-β2 and PcGABAA-β co-expression generated a chloride current that does not show desensitization. Muscimol reproduced the time course of GABA-evoked currents in all functional receptors, and picrotoxin blocked these currents; bicuculline did not block any of the recorded currents. Reverse transcription polymerae chain reaction (RT-PCR) amplifications and FISH revealed that PcGABAA-α and PcGABAA-β2 are predominantly expressed in the crayfish nervous system. Altogether, these findings provide the first evidence of a neural GABA-gated cationic channel in the crayfish, increasing our understanding of the role of these new GABAA receptor subunits in native heteromeric receptors.
Collapse
Affiliation(s)
- Iván Uriel Valladares-Hernández
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Juan Manuel Hernández-Martínez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - José Miguel Cuaxospa
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | | | - Edith Sánchez-Jaramillo
- Laboratorio de Neuroendocrinología Molecular, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Juan Manuel Arias
- Proyecto de Neurociencias, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Ubaldo García
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
22
|
Kan C, Ullah A, Dang S, Xue H. Modular Structure and Polymerization Status of GABA A Receptors Illustrated with EM Analysis and AlphaFold2 Prediction. Int J Mol Sci 2024; 25:10142. [PMID: 39337627 PMCID: PMC11432007 DOI: 10.3390/ijms251810142] [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: 06/29/2024] [Revised: 08/31/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Type-A γ-aminobutyric acid (GABAA) receptors are channel proteins crucial to mediating neuronal balance in the central nervous system (CNS). The structure of GABAA receptors allows for multiple binding sites and is key to drug development. Yet the formation mechanism of the receptor's distinctive pentameric structure is still unknown. This study aims to investigate the role of three predominant subunits of the human GABAA receptor in the formation of protein pentamers. Through purifying and refolding the protein fragments of the GABAA receptor α1, β2, and γ2 subunits, the particle structures were visualised with negative staining electron microscopy (EM). To aid the analysis, AlphaFold2 was used to compare the structures. Results show that α1 and β2 subunit fragments successfully formed homo-oligomers, particularly homopentameric structures, while the predominant heteropentameric GABAA receptor was also replicated through the combination of the three subunits. However, homopentameric structures were not observed with the γ2 subunit proteins. A comparison of the AlphaFold2 predictions and the previously obtained cryo-EM structures presents new insights into the subunits' modular structure and polymerization status. By performing experimental and computational studies, a deeper understanding of the complex structure of GABAA receptors is provided. Hopefully, this study can pave the way to developing novel therapeutics for neuropsychiatric diseases.
Collapse
Affiliation(s)
| | | | | | - Hong Xue
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; (C.K.); (A.U.); (S.D.)
| |
Collapse
|
23
|
Bartos M, Gallegos CE, Mónaco N, Lencinas I, Dominguez S, Bras C, Del Carmen Esandi M, Bouzat C, Gumilar F. Developmental exposure to arsenic reduces anxiety levels and leads to a depressive-like behavior in female offspring rats: Molecular changes in the prefrontal cortex. Neurotoxicology 2024; 104:85-94. [PMID: 39079579 DOI: 10.1016/j.neuro.2024.07.013] [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: 12/27/2023] [Revised: 07/18/2024] [Accepted: 07/27/2024] [Indexed: 08/05/2024]
Abstract
Exposure to inorganic arsenic (iAs) detrimentally affects the structure and function of the central nervous system. In-utero and postnatal exposure to iAs has been connected to adverse effects on cognitive development. Therefore, this investigation explores neurobehavioral and neurochemical effects of 0.05 and 0.10 mg/L iAs exposure during gestation and lactation periods on 90-day-old female offspring rats. The assessment of anxiety- and depressive-like behaviors was conducted through the application of an elevated plus maze and a forced swim test. The neurochemical changes were evaluated in the prefrontal cortex (PFC) through the determination of enzyme activities and α1 GABAA subunit expression levels. Our findings revealed a notable impact of iAs exposure on anxiety and the induction of depressive-like behavior in 90-day-old female offspring. Furthermore, the antioxidant status within the PFC exhibited discernible alterations in exposed rats. Notably, the activities of acetylcholinesterase and glutamate pyruvate transaminase demonstrated an increase, while glutamate oxaloacetate transaminase activity displayed a decrease within the PFC due to the iAs treatment. Additionally, a distinct downregulation in the mRNA expression of the α1GABAA receptor was observed in this neuronal region. These findings strongly suggest that iAs exposure during early stages of rat development causes significant modifications in brain oxidative stress markers and perturbs the activity of enzymes associated with cholinergic and glutamatergic systems. In parallel, it elicits a discernible reduction in the level of GABA receptors within the PFC. These molecular alterations may play a role in the diminished anxiety levels and the depressive-like behavior outlined in the current investigation.
Collapse
Affiliation(s)
- Mariana Bartos
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR) Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca CP8000, Argentina
| | - Cristina E Gallegos
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR) Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca CP8000, Argentina
| | - Nina Mónaco
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR) Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca CP8000, Argentina
| | - Ileana Lencinas
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR) Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca CP8000, Argentina
| | - Sergio Dominguez
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR) Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca CP8000, Argentina
| | - Cristina Bras
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR) Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca CP8000, Argentina
| | - María Del Carmen Esandi
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-CONICET, Bahía Blanca, Buenos Aires 8000, Argentina
| | - Cecilia Bouzat
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-CONICET, Bahía Blanca, Buenos Aires 8000, Argentina
| | - Fernanda Gumilar
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR) Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca CP8000, Argentina.
| |
Collapse
|
24
|
Câmara JS, Perestrelo R, Ferreira R, Berenguer CV, Pereira JAM, Castilho PC. Plant-Derived Terpenoids: A Plethora of Bioactive Compounds with Several Health Functions and Industrial Applications-A Comprehensive Overview. Molecules 2024; 29:3861. [PMID: 39202940 PMCID: PMC11357518 DOI: 10.3390/molecules29163861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Terpenoids are a large class of natural secondary plant metabolites which are highly diverse in structure, formed from isoprene units (C-5), associated with a wide range of biological properties, including antioxidant, antimicrobial, anti-inflammatory, antiallergic, anticancer, antimetastatic, antiangiogenesis, and apoptosis induction, and are considered for potential application in the food, cosmetics, pharmaceutical, and medical industries. In plants, terpenoids exert a variety of basic functions in growth and development. This review gives an overview, highlighting the current knowledge of terpenoids and recent advances in our understanding of the organization, regulation, and diversification of core and specialized terpenoid metabolic pathways and addressing the most important functions of volatile and non-volatile specialized terpenoid metabolites in plants. A comprehensive description of different aspects of plant-derived terpenoids as a sustainable source of bioactive compounds, their biosynthetic pathway, the several biological properties attributed to these secondary metabolites associated with health-promoting effects, and their potential industrial applications in several fields will be provided, and emerging and green extraction methods will also be discussed. In addition, future research perspectives within this framework will be highlighted. Literature selection was carried out using the National Library of Medicine, PubMed, and international reference data for the period from 2010 to 2024 using the keyword "terpenoids". A total of 177,633 published papers were found, of which 196 original and review papers were included in this review according to the criteria of their scientific reliability, their completeness, and their relevance to the theme considered.
Collapse
Affiliation(s)
- José S. Câmara
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal; (R.P.); (R.F.); (C.V.B.); (J.A.M.P.); (P.C.C.)
- Departamento de Química, Faculdade de Ciências Exatas e da Engenharia da Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Rosa Perestrelo
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal; (R.P.); (R.F.); (C.V.B.); (J.A.M.P.); (P.C.C.)
| | - Rui Ferreira
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal; (R.P.); (R.F.); (C.V.B.); (J.A.M.P.); (P.C.C.)
| | - Cristina V. Berenguer
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal; (R.P.); (R.F.); (C.V.B.); (J.A.M.P.); (P.C.C.)
| | - Jorge A. M. Pereira
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal; (R.P.); (R.F.); (C.V.B.); (J.A.M.P.); (P.C.C.)
| | - Paula C. Castilho
- CQM—Centro de Química da Madeira, NPRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal; (R.P.); (R.F.); (C.V.B.); (J.A.M.P.); (P.C.C.)
- Departamento de Química, Faculdade de Ciências Exatas e da Engenharia da Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| |
Collapse
|
25
|
Roy MS, Sarkar BK, Kundu SK. Site specific binding pattern of benzodiazepines with GABA A receptor and related impact on their activities in the human body: An in silico method based study. Heliyon 2024; 10:e33929. [PMID: 39071685 PMCID: PMC11283148 DOI: 10.1016/j.heliyon.2024.e33929] [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: 07/08/2023] [Revised: 06/29/2024] [Accepted: 06/30/2024] [Indexed: 07/30/2024] Open
Abstract
Despite of being the drugs of the same therapeutic class (Benzodiazepines), each of them shows different actions prominently. It is commonly seen that Bromazepam, Clonazepam, and, Alprazolam are prescribed for the treatment of anxiety disorders, panic disorders, and phobias. On the other hand, Midazolam, Temazepam, Flurazepam, and Nitrazepam are indicated for the treatment of insomnia and Lorazepam is considered as a drug having anticonvulsant effects. As the mechanism of action is the same, there should be some differences in the binding patterns with the proteins that create differences in their impacts on the body. A deep screening of the binding patterns of the available Benzodiazepines in the market to the GABAA receptor will be beneficial to find out the responsible amino acids for being accountable for showing any specific action. This reveal will help design new molecules with the highest beneficial effect and lowest toxicity in the body. The in silico method provides the initial level of understanding regarding the binding patterns, performing in vitro and in vivo experiments will be more specific to claim the benefits of newly designed drugs.
Collapse
Affiliation(s)
- Matrika Saha Roy
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | | | | |
Collapse
|
26
|
Xie W, Donat A, Jiang S, Baranowsky A, Keller J. The emerging role of tranexamic acid and its principal target, plasminogen, in skeletal health. Acta Pharm Sin B 2024; 14:2869-2884. [PMID: 39027253 PMCID: PMC11252461 DOI: 10.1016/j.apsb.2024.03.033] [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: 11/22/2023] [Revised: 02/28/2024] [Accepted: 03/14/2024] [Indexed: 07/20/2024] Open
Abstract
The worldwide burden of skeletal diseases such as osteoporosis, degenerative joint disease and impaired fracture healing is steadily increasing. Tranexamic acid (TXA), a plasminogen inhibitor and anti-fibrinolytic agent, is used to reduce bleeding with high effectiveness and safety in major surgical procedures. With its widespread clinical application, the effects of TXA beyond anti-fibrinolysis have been noticed and prompted renewed interest in its use. Some clinical trials have characterized the effects of TXA on reducing postoperative infection rates and regulating immune responses in patients undergoing surgery. Also, several animal studies suggest potential therapeutic effects of TXA on skeletal diseases such as osteoporosis and fracture healing. Although a direct effect of TXA on the differentiation and function of bone cells in vitro was shown, few mechanisms of action have been reported. Here, we summarize recent findings of the effects of TXA on skeletal diseases and discuss the underlying plasminogen-dependent and -independent mechanisms related to bone metabolism and the immune response. We furthermore discuss potential novel indications for TXA application as a treatment strategy for skeletal diseases.
Collapse
Affiliation(s)
- Weixin Xie
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Antonia Donat
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Shan Jiang
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Anke Baranowsky
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| |
Collapse
|
27
|
Zhao J, Chen AQ, Ryu J, del Mármol J. Structural basis of odor sensing by insect heteromeric odorant receptors. Science 2024; 384:1460-1467. [PMID: 38870275 PMCID: PMC11235583 DOI: 10.1126/science.adn6384] [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: 12/21/2023] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Most insects, including human-targeting mosquitoes, detect odors through odorant-activated ion channel complexes consisting of a divergent odorant-binding subunit (OR) and a conserved co-receptor subunit (Orco). As a basis for understanding how odorants activate these heteromeric receptors, we report here cryo-electron microscopy structures of two different heteromeric odorant receptor complexes containing ORs from disease-vector mosquitos Aedes aegypti or Anopheles gambiae. These structures reveal an unexpected stoichiometry of one OR to three Orco subunits. Comparison of structures in odorant-bound and unbound states indicates that odorant binding to the sole OR subunit is sufficient to open the channel pore, suggesting a mechanism of OR activation and a conceptual framework for understanding evolution of insect odorant receptor sensitivity.
Collapse
Affiliation(s)
- Jiawei Zhao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School; Boston, 02115, USA
| | - Andy Q. Chen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School; Boston, 02115, USA
| | - Jaewook Ryu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School; Boston, 02115, USA
| | - Josefina del Mármol
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School; Boston, 02115, USA
- Howard Hughes Medical Institute; Boston, 02115, USA
| |
Collapse
|
28
|
Lu X, Shu HJ, Lambert PM, Benz A, Zorumski CF, Mennerick S. δ-containing GABA A receptors on parvalbumin interneurons modulate neuronal excitability and network dynamics in the mouse medial prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599033. [PMID: 38915641 PMCID: PMC11195237 DOI: 10.1101/2024.06.14.599033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
In medial prefrontal cortex (mPFC), fast-spiking parvalbumin (PV) interneurons regulate excitability and microcircuit oscillatory activity important for cognition. Although PV interneurons inhibit pyramidal neurons, they themselves express δ subunits of GABAA receptors important for slow inhibition. However, the specific contribution of δ-containing GABAA receptors to the function of PV interneurons in mPFC is unclear. We explored cellular, synaptic, and local-circuit activity in PV interneurons and pyramidal neurons in mouse mPFC after selectively deleting δ subunits in PV interneurons (cKO mice). In current-clamp recordings, cKO PV interneurons exhibited a higher frequency of action potentials and higher input resistance than wild type (WT) PV interneurons. Picrotoxin increased firing and GABA decreased firing in WT PV interneurons but not in cKO PV interneurons. The δ-preferring agonist THIP reduced spontaneous inhibitory postsynaptic currents in WT pyramidal neurons but not in cKO pyramidal neurons. In WT slices, depolarizing the network with 400 nM kainate increased firing of pyramidal neurons but had little effect on PV interneuron firing. By contrast, in cKO slices kainate recruited PV interneurons at the expense of pyramidal neurons. At the population level, kainate induced broadband increases in local field potentials in WT but not cKO slices. These results on cells and the network can be understood through increased excitability of cKO PV interneurons. In summary, our study demonstrates that δ-containing GABAA receptors in mPFC PV interneurons play a crucial role in regulating their excitability and the phasic inhibition of pyramidal neurons, elucidating intricate mechanisms governing cortical circuitry. Significance statement By selectively deleting δ-containing GABAA receptors in PV interneurons, we demonstrate the importance of these receptors on PV interneuron excitability, synaptic inhibition of pyramidal neurons, and circuit function.
Collapse
|
29
|
Misrani A, Tabassum S, Wang T, Huang H, Jiang J, Diao H, Zhao Y, Huang Z, Tan S, Long C, Yang L. Vibration-reduced anxiety-like behavior relies on ameliorating abnormalities of the somatosensory cortex and medial prefrontal cortex. Neural Regen Res 2024; 19:1351-1359. [PMID: 37905885 PMCID: PMC11467954 DOI: 10.4103/1673-5374.385840] [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: 02/07/2023] [Revised: 05/30/2023] [Accepted: 07/19/2023] [Indexed: 11/02/2023] Open
Abstract
Tibetan singing bowls emit low-frequency sounds and produce perceptible harmonic tones and vibrations through manual tapping. The sounds the singing bowls produce have been shown to enhance relaxation and reduce anxiety. However, the underlying mechanism remains unclear. In this study, we used chronic restraint stress or sleep deprivation to establish mouse models of anxiety that exhibit anxiety-like behaviors. We then supplied treatment with singing bowls in a bottomless cage placed on the top of a cushion. We found that unlike in humans, the combination of harmonic tones and vibrations did not improve anxiety-like behaviors in mice, while individual vibration components did. Additionally, the vibration of singing bowls increased the level of N-methyl-D-aspartate receptor 1 in the somatosensory cortex and prefrontal cortex of the mice, decreased the level of γ-aminobutyric acid A (GABA) receptor α 1 subtype, reduced the level of CaMKII in the prefrontal cortex, and increased the number of GABAergic interneurons. At the same time, electrophysiological tests showed that the vibration of singing bowls significantly reduced the abnormal low-frequency gamma oscillation peak frequency in the medial prefrontal cortex caused by stress restraint pressure and sleep deprivation. Results from this study indicate that the vibration of singing bowls can alleviate anxiety-like behaviors by reducing abnormal molecular and electrophysiological events in somatosensory and medial prefrontal cortex.
Collapse
Affiliation(s)
- Afzal Misrani
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Sidra Tabassum
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Tintin Wang
- Guangzhou Hongai Cultural Development, Inc., Guangzhou, Guangdong Province, China
- Yinguo Health Management Team, Guangzhou, Guangdong Province, China
| | - Huixian Huang
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Jinxiang Jiang
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Hongjun Diao
- Guangzhou Hongai Cultural Development, Inc., Guangzhou, Guangdong Province, China
- Yinguo Health Management Team, Guangzhou, Guangdong Province, China
| | - Yanping Zhao
- College of Biophotonics, South China Normal University, Guangzhou, Guangdong Province, China
| | - Zhen Huang
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
| | - Shaohua Tan
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
| | - Cheng Long
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
| |
Collapse
|
30
|
Curry RN, Ma Q, McDonald MF, Ko Y, Srivastava S, Chin PS, He P, Lozzi B, Athukuri P, Jing J, Wang S, Harmanci AO, Arenkiel B, Jiang X, Deneen B, Rao G, Harmanci AS. Integrated electrophysiological and genomic profiles of single cells reveal spiking tumor cells in human glioma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583026. [PMID: 38496434 PMCID: PMC10942290 DOI: 10.1101/2024.03.02.583026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Prior studies have described the complex interplay that exists between glioma cells and neurons, however, the electrophysiological properties endogenous to tumor cells remain obscure. To address this, we employed Patch-sequencing on human glioma specimens and found that one third of patched cells in IDH mutant (IDH mut ) tumors demonstrate properties of both neurons and glia by firing single, short action potentials. To define these hybrid cells (HCs) and discern if they are tumor in origin, we developed a computational tool, Single Cell Rule Association Mining (SCRAM), to annotate each cell individually. SCRAM revealed that HCs represent tumor and non-tumor cells that feature GABAergic neuron and oligodendrocyte precursor cell signatures. These studies are the first to characterize the combined electrophysiological and molecular properties of human glioma cells and describe a new cell type in human glioma with unique electrophysiological and transcriptomic properties that are likely also present in the non-tumor mammalian brain.
Collapse
|
31
|
Bukanova JV, Solntseva EI, Skrebitsky VG. Factors promoting the release of picrotoxin from the trap in the GABA(A) receptor pore. Neurochem Int 2024; 175:105703. [PMID: 38395151 DOI: 10.1016/j.neuint.2024.105703] [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: 12/05/2023] [Revised: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 02/25/2024]
Abstract
Picrotoxin (PTX), a convulsant of plant origin, has been used in many studies as research tool. PTX is the open channel blocker of the GABAA receptor (GABAAR). Being in the pore, PTX initiates transfer of the channel to the closed state and thus it falls into the "trap". The consequence of this PTX trapping is so-called aftereffect, i.e. continuation of the blockade of the GABA-induced chloride current (IGABA) after removal of PTX from the external solution. The present work shows that the positive allosteric modulators (PAMs) of the GABAA receptor, allopregnanolone (Allo) and zolpidem (Zolp) as well as a high concentration of GABA shortened the PTX aftereffect. Experiments were carried out on isolated Purkinje neurons of the rat cerebellum using the whole-cell patch-clamp method. IGABA was induced by applications of 5 μM GABA (EC30) for 1 s with 30 s intervals. 50 μM PTX completely blocked IGABA, and recovery upon PTX washout occurred with a time constant (τrec) of 20.2 min. 1 μM Allo reduced the blocking effect of PTX by 30% and accelerated the recovery of IGABA by almost 10 times (τrec = 2.4 min). 0.5 μM Zolp did not change the IGABA block in the presence of PTX but accelerated the recovery of IGABA by more than 3 times (τrec = 5.6 min). Increasing the GABA concentration to 20 μM did not change the blocking effect of PTX, but accelerated recovery by 6 times (τrec = 3.3 min). The mechanism of the shortening of the PTX aftereffect is presumably the expansion of the GABAAR pore in the presence of PAMs and a high concentration of the agonist and, as a consequence, the escape of PTX from the "trap". The work describes new pharmacological properties of Allo and Zolp.
Collapse
Affiliation(s)
- Julia V Bukanova
- Brain Research Institute, Research Center of Neurology, Moscow, Russia
| | - Elena I Solntseva
- Brain Research Institute, Research Center of Neurology, Moscow, Russia.
| | | |
Collapse
|
32
|
Colucci Cante R, Nigro F, Passannanti F, Lentini G, Gallo M, Nigro R, Budelli AL. Gut health benefits and associated systemic effects provided by functional components from the fermentation of natural matrices. Compr Rev Food Sci Food Saf 2024; 23:e13356. [PMID: 38767859 DOI: 10.1111/1541-4337.13356] [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: 10/09/2023] [Revised: 02/26/2024] [Accepted: 04/06/2024] [Indexed: 05/22/2024]
Abstract
Recently, the role of the gut microbiota in metabolic health, immunity, behavioral balance, longevity, and intestine comfort has been the object of several studies from scientific communities. They were encouraged by a growing interest from food industries and consumers toward novel fermented ingredients and formulations with powerful biological effects, such as pre, pro, and postbiotic products. Depending on the selected strains, the operating conditions, the addition of suitable reagents or enzymes, the equipment, and the reactor configurations, functional compounds with high bioactivity, such as short-chain fatty acids, gamma-aminobutyric acid, bioactive peptides, and serotonin, can be enhanced and/or produced through fermentation of several vegetable matrices. Otherwise, their formation can also be promoted directly in the gut after the dietary intake of fermented foods: In this case, fermentation will aim to increase the content of precursor substances, such as indigestible fibers, polyphenols, some amino acids, and resistant starch, which can be potentially metabolized by endogenous gut microorganisms and converted in healthy molecules. This review provides an overview of the main functional components currently investigated in literature and the associated gut health benefits. The current state of the art about fermentation technology as a promising functionalization tool to promote the direct or indirect formation of gut-health-enhancing components was deepened, highlighting the importance of optimizing microorganism selection, system setups, and process conditions according to the target compound of interest. The collected data suggested the possibility of gaining novel functional food ingredients or products rich in functional molecules through fermentation without performing additional extraction and purification stages, which are needed when conventional culture broths are used.
Collapse
Affiliation(s)
- Rosa Colucci Cante
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
- Department of Industrial Engineering, University of Niccolò Cusano, Rome, Italy
| | - Federica Nigro
- I. T. P. Innovation and Technology Provider S.r.l., Naples, Italy
| | - Francesca Passannanti
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
- I. T. P. Innovation and Technology Provider S.r.l., Naples, Italy
| | - Giulia Lentini
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
| | - Marianna Gallo
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
- Department of Industrial Engineering, University of Niccolò Cusano, Rome, Italy
- I. T. P. Innovation and Technology Provider S.r.l., Naples, Italy
| | - Roberto Nigro
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
33
|
Wright NJD. A review of the direct targets of the cannabinoids cannabidiol, Δ9-tetrahydrocannabinol, N-arachidonoylethanolamine and 2-arachidonoylglycerol. AIMS Neurosci 2024; 11:144-165. [PMID: 38988890 PMCID: PMC11230856 DOI: 10.3934/neuroscience.2024009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 07/12/2024] Open
Abstract
Marijuana has been used by humans for thousands of years for both medicinal and recreational purposes. This included the treatment of pain, inflammation, seizures, and nausea. In the 1960s, the structure of the principal psychoactive ingredient Δ9-tetrahydrocannabinol was determined, and over the next few decades, two cannabinoid receptors were characterized along with the human endocannabinoid system and what it affects. This includes metabolism, the cardiovascular and reproductive systems, and it is involved in such conditions as inflammation, cancer, glaucoma, and liver and musculoskeletal disorders. In the central nervous system, the endocannabinoid system has been linked to appetite, learning, memory, and conditions such as depression, anxiety, schizophrenia, stroke, multiple sclerosis, neurodegeneration, addiction, and epilepsy. It was the profound effectiveness of cannabidiol, a non-psychoactive ingredient of marijuana, to relieve the symptoms of Dravet syndrome, a severe form of childhood epilepsy, that recently helped spur marijuana research. This has helped substantially to change society's attitude towards this potential source of useful drugs. However, research has also revealed that the actions of endocannabinoids, such as anandamide and 2-arachidonoylglycerol, and the phytocannabinoids, tetrahydrocannabinol and cannabidiol, were not just due to interactions with the two cannabinoid receptors but by acting directly on many other targets including various G-protein receptors and cation channels, such as the transient receptor potential channels for example. This mini-review attempts to survey the effects of these 4 important cannabinoids on these currently identified targets.
Collapse
|
34
|
Tateiwa H, Evers AS. Neurosteroids and their potential as a safer class of general anesthetics. J Anesth 2024; 38:261-274. [PMID: 38252143 PMCID: PMC10954990 DOI: 10.1007/s00540-023-03291-4] [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: 10/31/2023] [Accepted: 11/25/2023] [Indexed: 01/23/2024]
Abstract
Neurosteroids (NS) are a class of steroids that are synthesized within the central nervous system (CNS). Various NS can either enhance or inhibit CNS excitability and they play important biological roles in brain development, brain function and as mediators of mood. One class of NS, 3α-hydroxy-pregnane steroids such as allopregnanolone (AlloP) or pregnanolone (Preg), inhibits neuronal excitability; these endogenous NS and their analogues have been therapeutically applied as anti-depressants, anti-epileptics and general anesthetics. While NS have many favorable properties as anesthetics (e.g. rapid onset, rapid recovery, minimal cardiorespiratory depression, neuroprotection), they are not currently in clinical use, largely due to problems with formulation. Recent advances in understanding NS mechanisms of action and improved formulations have rekindled interest in development of NS as sedatives and anesthetics. In this review, the synthesis of NS, and their mechanism of action will be reviewed with specific emphasis on their binding sites and actions on γ-aminobutyric acid type A (GABAA) receptors. The potential advantages of NS analogues as sedative and anesthetic agents will be discussed.
Collapse
Affiliation(s)
- Hiroki Tateiwa
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kochi, Japan
| | - Alex S Evers
- Department of Anesthesiology, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO, 63110, USA.
| |
Collapse
|
35
|
Witkin JM, Shafique H, Cerne R, Smith JL, Marini AM, Lipsky RH, Delery E. Mechanistic and therapeutic relationships of traumatic brain injury and γ-amino-butyric acid (GABA). Pharmacol Ther 2024; 256:108609. [PMID: 38369062 DOI: 10.1016/j.pharmthera.2024.108609] [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: 10/28/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Traumatic brain injury (TBI) is a highly prevalent medical condition for which no medications specific for the prophylaxis or treatment of the condition as a whole exist. The spectrum of symptoms includes coma, headache, seizures, cognitive impairment, depression, and anxiety. Although it has been known for years that the inhibitory neurotransmitter γ-amino-butyric acid (GABA) is involved in TBI, no novel therapeutics based upon this mechanism have been introduced into clinical practice. We review the neuroanatomical, neurophysiological, neurochemical, and neuropharmacological relationships of GABA neurotransmission to TBI with a view toward new potential GABA-based medicines. The long-standing idea that excitatory and inhibitory (GABA and others) balances are disrupted by TBI is supported by the experimental data but has failed to invent novel methods of restoring this balance. The slow progress in advancing new treatments is due to the complexity of the disorder that encompasses multiple dynamically interacting biological processes including hemodynamic and metabolic systems, neurodegeneration and neurogenesis, major disruptions in neural networks and axons, frank brain lesions, and a multitude of symptoms that have differential neuronal and neurohormonal regulatory mechanisms. Although the current and ongoing clinical studies include GABAergic drugs, no novel GABA compounds are being explored. It is suggested that filling the gap in understanding the roles played by specific GABAA receptor configurations within specific neuronal circuits could help define new therapeutic approaches. Further research into the temporal and spatial delivery of GABA modulators should also be useful. Along with GABA modulation, research into the sequencing of GABA and non-GABA treatments will be needed.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | | | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA; Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA
| | - Ann M Marini
- Department of Neurology, Program in Neuroscience, and Molecular and Cellular Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert H Lipsky
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Elizabeth Delery
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA.
| |
Collapse
|
36
|
Chang Y, Xie X, Liu Y, Liu M, Zhang H. Exploring clinical applications and long-term effectiveness of benzodiazepines: An integrated perspective on mechanisms, imaging, and personalized medicine. Biomed Pharmacother 2024; 173:116329. [PMID: 38401518 DOI: 10.1016/j.biopha.2024.116329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024] Open
Abstract
Benzodiazepines have been long-established treatments for various conditions, including anxiety disorders and insomnia. Recent FDA warnings emphasize the risks of misuse and dependence associated with benzodiazepines. This article highlights their benefits and potential drawbacks from various perspectives. It achieves this by explaining how benzodiazepines work in terms of neuroendocrinology, immunomodulation, sleep, anxiety, cognition, and addiction, ultimately improving their clinical effectiveness. Benzodiazepines play a regulatory role in the HPA axis and impact various systems, including neuropeptide Y and cholecystokinin. Benzodiazepines can facilitate sleep-dependent memory consolidation by promoting spindle wave activity, but they can also lead to memory deficits in older individuals due to reduced slow-wave sleep. The cognitive effects of chronic benzodiazepines use remain uncertain; however, no adverse findings have been reported in clinical imaging studies. This article aims to comprehensively review the evidence on benzodiazepines therapy, emphasizing the need for more clinical studies, especially regarding long-term benzodiazepines use.
Collapse
Affiliation(s)
- Yiheng Chang
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xueting Xie
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yudan Liu
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Meichen Liu
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Huimin Zhang
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
37
|
Guo Y, Kang Y, Bai W, Liu Q, Zhang R, Wang Y, Wang C. Perinatal exposure to bisphenol A impairs cognitive function via the gamma-aminobutyric acid signaling pathway in male rat offspring. ENVIRONMENTAL TOXICOLOGY 2024; 39:1235-1244. [PMID: 37926988 DOI: 10.1002/tox.24007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/17/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
Bisphenol A (BPA) is a common synthetic endocrine disruptor that can be utilized in the fabrication of materials such as polycarbonates and epoxy resins. Numerous studies have linked BPA to learning and memory problems, although the precise mechanism remains unknown. Gamma-aminobutyric acid (GABA) is the most abundant inhibitory neurotransmitter in the vertebrate central nervous system, and it is intimately related to learning and memory. This study aims to evaluate whether altered cognitive behavior involves the GABA signaling pathway in male offspring of rats exposed to BPA during the prenatal and early postnatal periods. Pregnant rats were orally given BPA (0, 0.04, 0.4, and 4 mg/kg body weight (BW)/day) from the first day of pregnancy to the 21st day of breastfeeding. Three-week-old male rat offspring were selected for an open-field experiment and a new object recognition experiment to evaluate the effect of BPA exposure on cognitive behavior. Furthermore, the role of GABA signaling markers in the cognition affected by BPA was investigated at the molecular level using western blotting and real-time polymerase chain reaction (RT-PCR). The research demonstrated that BPA exposure impacted the behavior and memory of male rat offspring and elevated the expression of glutamic acid decarboxylase 67 (GAD67), GABA type A receptors subunit (GABAARα1), and GABA vesicle transporter (VGAT) in the hippocampus while decreasing the expression levels of GABA transaminase (GABA-T) and GABA transporter 1 (GAT-1). These findings indicate that the alteration in the expression of GABA signaling molecules may be one of the molecular mechanisms by which perinatal exposure to BPA leads to decreased learning and memory in male rat offspring.
Collapse
Affiliation(s)
- Yi Guo
- College of Health Public, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yuxin Kang
- College of Health Public, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Wenjie Bai
- College of Health Public, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Qiling Liu
- College of Health Public, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Rongqiang Zhang
- College of Health Public, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yuxin Wang
- College of Health Public, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Chong Wang
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
38
|
Richardson RJ, Petrou S, Bryson A. Established and emerging GABA A receptor pharmacotherapy for epilepsy. Front Pharmacol 2024; 15:1341472. [PMID: 38449810 PMCID: PMC10915249 DOI: 10.3389/fphar.2024.1341472] [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: 11/20/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
Drugs that modulate the GABAA receptor are widely used in clinical practice for both the long-term management of epilepsy and emergency seizure control. In addition to older medications that have well-defined roles for the treatment of epilepsy, recent discoveries into the structure and function of the GABAA receptor have led to the development of newer compounds designed to maximise therapeutic benefit whilst minimising adverse effects, and whose position within the epilepsy pharmacologic armamentarium is still emerging. Drugs that modulate the GABAA receptor will remain a cornerstone of epilepsy management for the foreseeable future and, in this article, we provide an overview of the mechanisms and clinical efficacy of both established and emerging pharmacotherapies.
Collapse
Affiliation(s)
- Robert J. Richardson
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Austin Health, Heidelberg, VIC, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Praxis Precision Medicines, Boston, MA, United States
| | - Alexander Bryson
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Austin Health, Heidelberg, VIC, Australia
- Department of Neurology, Eastern Health, Melbourne, VIC, Australia
| |
Collapse
|
39
|
Ali DN, Ali HM, Lopez MR, Kang S, Choi DS. Astrocytic GABAergic Regulation in Alcohol Use and Major Depressive Disorders. Cells 2024; 13:318. [PMID: 38391931 PMCID: PMC10887002 DOI: 10.3390/cells13040318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system (CNS). Most GABAergic neurons synthesize GABA from glutamate and release it in the synaptic cleft in the CNS. However, astrocytes can also synthesize and release GABA, activating GABA receptors in the neighboring neurons in physiological and pathological conditions. As the primary homeostatic glial cells in the brain, astrocytes play a crucial role in regulating GABA homeostasis and synaptic neurotransmission. Accumulating evidence demonstrates that astrocytic GABA dysregulation is implicated in psychiatric disorders, including alcohol use disorder (AUD) and major depressive disorder (MDD), the most prevalent co-occurring psychiatric disorders. Several current medications and emerging pharmacological agents targeting GABA levels are in clinical trials for treating AUD and MDD. This review offers a concise summary of the role of astrocytic GABA regulation in AUD and MDD. We also provide an overview of the current understanding and areas of debate regarding the mechanisms by which astrocytes regulate GABA in the CNS and their potential significance in the molecular basis of AUD and MDD, paving the way toward future research directions and potential therapeutic target areas within this field.
Collapse
Affiliation(s)
- Dina N. Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Hossam M. Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Matthew R. Lopez
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
- Neuroscience Program, Rochester, MN 55905, USA
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
40
|
de Menezes JFS, Sá Pires Silva AM, Aparecida Faria de Almeida E, da Silva AF, Morais Bomfim De Lima J, da Silva AW, Ferreira MKA, de Menezes JESA, Dos Santos HS, Marinho ES, Marinho GS, Marques da Fonseca A. Synthesis and anxiolytic effect of europium metallic complex containing lapachol [Eu(DBM) 3. LAP] in adult zebrafish through serotonergic neurotransmission: in vivo and in silico approach. J Biomol Struct Dyn 2024; 42:1280-1292. [PMID: 37029769 DOI: 10.1080/07391102.2023.2199087] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/29/2023] [Indexed: 04/09/2023]
Abstract
Anxiety-related mental health problems are estimated at 3.6% globally, benzodiazepines (BZDs) are the class of drugs indicated for the treatment of anxiety, including lorazepam and diazepam. However, concerns have been raised about the short- and long-term risks associated with BZDs. Therefore, despite anxiolytic and antidepressant drugs, there is a need to develop more effective pharmacotherapies with fewer side effects than existing drugs. The present work reported the synthesis, anxiolytic activity, mechanism of action in Adult Zebrafish (Danio rerio) and in silico study of a europium metallic complex with Lapachol, [Eu(DBM)3. LAP]. Each animal (n = 6/group) was treated intraperitoneally (i.p.; 20 µL) with the synthesized complex (4, 20 and 40 mg/Kg) and with the vehicle (DMSO 3%; 20 µL), being submitted to the tests of locomotor activity and 96h acute toxicity. The light/dark test was also performed, and the serotonergic mechanism (5-HT) was evaluated through the antagonists of the 5-HTR1, 5-HTR2A/2C and 5-HTR3A/3B receptors. The complex was characterized using spectrometric techniques, and the anxiolytic effect of complex may be involved the neuromodulation of receptors 5-HT3A/3B, since the pre-treatment with pizotifen and cyproheptadine did not block the anxiolytic effect of [Eu(DBM)3. LAP], unlike fluoxetine had its anxiolytic effect reversed. In addition, molecular docking showed interaction between the [Eu(DBM)3. LAP] and 5HT3A receptor with binding energy -7.8 kcal/mol and the ADMET study showed that complex has low toxic risk. It is expected that the beginning of this study will allow the application of the new anxiolytic drugs, given the pharmacological potential of the lapachol complex.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jorge Fernando Silva de Menezes
- Center for Teacher Training, Federal University of Recôncavo da Bahia, Amargosa, Bahia, Brazil
- INCT - Energia e Meio Ambiente, UFBA, Rua Barão de Jeremoabo, Salvador, Bahia, Brazil
| | | | | | - Ananias Freire da Silva
- Postgraduate Program in Energy and Environment - PGEA, Institute of Engineering and Sustainable Development, University of International Integration of Afro-Brazilian Lusofonia, Acarape, Ceará, Brazil
| | | | | | | | | | - Hélcio Silva Dos Santos
- State University of Ceará, Graduate Program in Natural Sciences, Fortaleza, Ceará, Brazil
- State University of Vale do Acaraú, Chemistry Course, Sobral, Ceará, Brazil
| | - Emmanuel Silva Marinho
- State University of Ceará, Graduate Program in Natural Sciences, Fortaleza, Ceará, Brazil
- Degree Course in Computer Science, Ceará State University, Fortaleza, Ceará, Brazil
| | | | - Aluísio Marques da Fonseca
- Postgraduate Program in Energy and Environment - PGEA, Institute of Engineering and Sustainable Development, University of International Integration of Afro-Brazilian Lusofonia, Acarape, Ceará, Brazil
| |
Collapse
|
41
|
Platonov M, Maximyuk O, Rayevsky A, Iegorova O, Hurmach V, Holota Y, Bulgakov E, Cherninskyi A, Karpov P, Ryabukhin S, Krishtal O, Volochnyuk D. Integrated workflow for the identification of new GABA A R positive allosteric modulators based on the in silico screening with further in vitro validation. Case study using Enamine's stock chemical space. Mol Inform 2024; 43:e202300156. [PMID: 37964718 DOI: 10.1002/minf.202300156] [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: 06/11/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/16/2023]
Abstract
Numerous studies reported an association between GABAA R subunit genes and epilepsy, eating disorders, autism spectrum disorders, neurodevelopmental disorders, and bipolar disorders. This study was aimed to find some potential positive allosteric modulators and was performed by combining the in silico approach with further in vitro evaluation of its real activity. We started from the GABAA R-diazepam complexes and assembled a lipid embedded protein ensemble to refine it via molecular dynamics (MD) simulation. Then we focused on the interaction of α1β2γ2 with some Z-drugs (non-benzodiazepine compounds) using an Induced Fit Docking (IFD) into the relaxed binding site to generate a pharmacophore model. The pharmacophore model was validated with a reference set and applied to decrease the pre-filtered Enamine database before the main docking procedure. Finally, we succeeded in identifying a set of compounds, which met all features of the docking model. The aqueous solubility and stability of these compounds in mouse plasma were assessed. Then they were tested for the biological activity using the rat Purkinje neurons and CHO cells with heterologously expressed human α1β2γ2 GABAA receptors. Whole-cell patch clamp recordings were used to reveal the GABA induced currents. Our study represents a convenient and tunable model for the discovery of novel positive allosteric modulators of GABAA receptors. A High-throughput virtual screening of the largest available database of chemical compounds resulted in the selection of 23 compounds. Further electrophysiological tests allowed us to determine a set of 3 the most outstanding active compounds. Considering the structural features of leader compounds, the study can develop into the MedChem project soon.
Collapse
Affiliation(s)
- Maksym Platonov
- Institute of molecular biology and genetics, Natl. Academy of Sciences of Ukraine, Zabolotnogo Str., 150, Kyiv, 03143, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
| | - Oleksandr Maximyuk
- Bogomoletz Institute of Physiology, Natl. Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024, Kyiv, Ukraine
| | - Alexey Rayevsky
- Institute of molecular biology and genetics, Natl. Academy of Sciences of Ukraine, Zabolotnogo Str., 150, Kyiv, 03143, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
- Institute of Food Biotechnology and Genomics, Natl. Academy of Sciences of Ukraine, Osypovskoho Str., 2 A, Kyiv, 04123, Ukraine
| | - Olena Iegorova
- Bogomoletz Institute of Physiology, Natl. Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024, Kyiv, Ukraine
| | - Vasyl Hurmach
- Institute of molecular biology and genetics, Natl. Academy of Sciences of Ukraine, Zabolotnogo Str., 150, Kyiv, 03143, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
| | - Yuliia Holota
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
| | - Elijah Bulgakov
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
- Institute of Food Biotechnology and Genomics, Natl. Academy of Sciences of Ukraine, Osypovskoho Str., 2 A, Kyiv, 04123, Ukraine
| | - Andrii Cherninskyi
- Bogomoletz Institute of Physiology, Natl. Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024, Kyiv, Ukraine
| | - Pavel Karpov
- Institute of Food Biotechnology and Genomics, Natl. Academy of Sciences of Ukraine, Osypovskoho Str., 2 A, Kyiv, 04123, Ukraine
| | - Sergey Ryabukhin
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
- Institute of High Technologies, Taras Shevchenko National University of Kyiv., Glushkova Ave, 03022, Kyiv, Ukraine
- Institute of organic chemistry NAS of Ukraine, 5 Murmanska Str., 02660, Kyiv, Ukraine
| | - Oleg Krishtal
- Bogomoletz Institute of Physiology, Natl. Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024, Kyiv, Ukraine
| | - Dmitriy Volochnyuk
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
- Institute of High Technologies, Taras Shevchenko National University of Kyiv., Glushkova Ave, 03022, Kyiv, Ukraine
- Institute of organic chemistry NAS of Ukraine, 5 Murmanska Str., 02660, Kyiv, Ukraine
| |
Collapse
|
42
|
Menzikov SA, Zaichenko DM, Moskovtsev AA, Morozov SG, Kubatiev AA. Phenols and GABA A receptors: from structure and molecular mechanisms action to neuropsychiatric sequelae. Front Pharmacol 2024; 15:1272534. [PMID: 38303988 PMCID: PMC10831359 DOI: 10.3389/fphar.2024.1272534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) are members of the pentameric ligand-gated ion channel (pLGIC) family, which are widespread throughout the invertebrate and vertebrate central nervous system. GABAARs are engaged in short-term changes of the neuronal concentrations of chloride (Cl-) and bicarbonate (HCO3 -) ions by their passive permeability through the ion channel pore. GABAARs are regulated by various structurally diverse phenolic substances ranging from simple phenols to complex polyphenols. The wide chemical and structural variability of phenols suggest similar and different binding sites on GABAARs, allowing them to manifest themselves as activators, inhibitors, or allosteric ligands of GABAAR function. Interest in phenols is associated with their great potential for GABAAR modulation, but also with their subsequent negative or positive role in neurological and psychiatric disorders. This review focuses on the GABAergic deficit hypotheses during neurological and psychiatric disorders induced by various phenols. We summarize the structure-activity relationship of general phenol groups concerning their differential roles in the manifestation of neuropsychiatric symptoms. We describe and analyze the role of GABAAR subunits in manifesting various neuropathologies and the molecular mechanisms underlying their modulation by phenols. Finally, we discuss how phenol drugs can modulate GABAAR activity via desensitization and resensitization. We also demonstrate a novel pharmacological approach to treat neuropsychiatric disorders via regulation of receptor phosphorylation/dephosphorylation.
Collapse
|
43
|
Lebovich M, Lora MA, Gracia-David J, Andrews LB. Genetic Circuits for Feedback Control of Gamma-Aminobutyric Acid Biosynthesis in Probiotic Escherichia coli Nissle 1917. Metabolites 2024; 14:44. [PMID: 38248847 PMCID: PMC10819706 DOI: 10.3390/metabo14010044] [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: 12/01/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024] Open
Abstract
Engineered microorganisms such as the probiotic strain Escherichia coli Nissle 1917 (EcN) offer a strategy to sense and modulate the concentration of metabolites or therapeutics in the gastrointestinal tract. Here, we present an approach to regulate the production of the depression-associated metabolite gamma-aminobutyric acid (GABA) in EcN using genetic circuits that implement negative feedback. We engineered EcN to produce GABA by overexpressing glutamate decarboxylase and applied an intracellular GABA biosensor to identify growth conditions that improve GABA biosynthesis. We next employed characterized genetically encoded NOT gates to construct genetic circuits with layered feedback to control the rate of GABA biosynthesis and the concentration of GABA produced. Looking ahead, this approach may be utilized to design feedback control of microbial metabolite biosynthesis to achieve designable smart microbes that act as living therapeutics.
Collapse
Affiliation(s)
- Matthew Lebovich
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Biotechnology Training Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Marcos A. Lora
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jared Gracia-David
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Lauren B. Andrews
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Biotechnology Training Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
44
|
Thompson SM. Modulators of GABA A receptor-mediated inhibition in the treatment of neuropsychiatric disorders: past, present, and future. Neuropsychopharmacology 2024; 49:83-95. [PMID: 37709943 PMCID: PMC10700661 DOI: 10.1038/s41386-023-01728-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/14/2023] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
The predominant inhibitory neurotransmitter in the brain, γ-aminobutyric acid (GABA), acts at ionotropic GABAA receptors to counterbalance excitation and regulate neuronal firing. GABAA receptors are heteropentameric channels comprised from subunits derived from 19 different genes. GABAA receptors have one of the richest and well-developed pharmacologies of any therapeutic drug target, including agonists, antagonists, and positive and negative allosteric modulators (PAMs, NAMs). Currently used PAMs include benzodiazepine sedatives and anxiolytics, barbiturates, endogenous and synthetic neurosteroids, and general anesthetics. In this article, I will review evidence that these drugs act at several distinct binding sites and how they can be used to alter the balance between excitation and inhibition. I will also summarize existing literature regarding (1) evidence that changes in GABAergic inhibition play a causative role in major depression, anxiety, postpartum depression, premenstrual dysphoric disorder, and schizophrenia and (2) whether and how GABAergic drugs exert beneficial effects in these conditions, focusing on human studies where possible. Where these classical therapeutics have failed to exert benefits, I will describe recent advances in clinical and preclinical drug development. I will also highlight opportunities to advance a generation of GABAergic therapeutics, such as development of subunit-selective PAMs and NAMs, that are engendering hope for novel tools to treat these devastating conditions.
Collapse
Affiliation(s)
- Scott M Thompson
- Center for Novel Therapeutics, Department of Psychiatry, University of Colorado School of Medicine, 12700 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
45
|
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
|
46
|
Maguire JL, Mennerick S. Neurosteroids: mechanistic considerations and clinical prospects. Neuropsychopharmacology 2024; 49:73-82. [PMID: 37369775 PMCID: PMC10700537 DOI: 10.1038/s41386-023-01626-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/15/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2023]
Abstract
Like other classes of treatments described in this issue's section, neuroactive steroids have been studied for decades but have risen as a new class of rapid-acting, durable antidepressants with a distinct mechanism of action from previous antidepressant treatments and from other compounds covered in this issue. Neuroactive steroids are natural derivatives of progesterone but are proving effective as exogenous treatments. The best understood mechanism is that of positive allosteric modulation of GABAA receptors, where subunit selectivity may promote their profile of action. Mechanistically, there is some reason to think that neuroactive steroids may separate themselves from liabilities of other GABA modulators, although research is ongoing. It is also possible that intracellular targets, including inflammatory pathways, may be relevant to beneficial actions. Strengths and opportunities for further development include exploiting non-GABAergic targets, structural analogs, enzymatic production of natural steroids, precursor loading, and novel formulations. The molecular mechanisms of behavioral effects are not fully understood, but study of brain network states involved in emotional processing demonstrate a robust influence on affective states not evident with at least some other GABAergic drugs including benzodiazepines. Ongoing studies with neuroactive steroids will further elucidate the brain and behavioral effects of these compounds as well as likely underpinnings of disease.
Collapse
Affiliation(s)
- Jamie L Maguire
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Steven Mennerick
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., St. Louis, MO, 63110, USA.
| |
Collapse
|
47
|
Masciocchi S, Businaro P, Scaranzin S, Morandi C, Franciotta D, Gastaldi M. General features, pathogenesis, and laboratory diagnostics of autoimmune encephalitis. Crit Rev Clin Lab Sci 2024; 61:45-69. [PMID: 37777038 DOI: 10.1080/10408363.2023.2247482] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/09/2023] [Indexed: 10/02/2023]
Abstract
Autoimmune encephalitis (AE) is a group of inflammatory conditions that can associate with the presence of antibodies directed to neuronal intracellular, or cell surface antigens. These disorders are increasingly recognized as an important differential diagnosis of infectious encephalitis and of other common neuropsychiatric conditions. Autoantibody diagnostics plays a pivotal role for accurate diagnosis of AE, which is of utmost importance for the prompt recognition and early treatment. Several AE subgroups can be identified, either according to the prominent clinical phenotype, presence of a concomitant tumor, or type of neuronal autoantibody, and recent diagnostic criteria have provided important insights into AE classification. Antibodies to neuronal intracellular antigens typically associate with paraneoplastic neurological syndromes and poor prognosis, whereas antibodies to synaptic/neuronal cell surface antigens characterize many AE subtypes that associate with tumors less frequently, and that are often immunotherapy-responsive. In addition to the general features of AE, we review current knowledge on the pathogenic mechanisms underlying these disorders, focusing mainly on the potential role of neuronal antibodies in the most frequent conditions, and highlight current theories and controversies. Then, we dissect the crucial aspects of the laboratory diagnostics of neuronal antibodies, which represents an actual challenge for both pathologists and neurologists. Indeed, this diagnostics entails technical difficulties, along with particularly interesting novel features and pitfalls. The novelties especially apply to the wide range of assays used, including specific tissue-based and cell-based assays. These assays can be developed in-house, usually in specialized laboratories, or are commercially available. They are widely used in clinical immunology and in clinical chemistry laboratories, with relevant differences in analytic performance. Indeed, several data indicate that in-house assays could perform better than commercial kits, notwithstanding that the former are based on non-standardized protocols. Moreover, they need expertise and laboratory facilities usually unavailable in clinical chemistry laboratories. Together with the data of the literature, we critically evaluate the analytical performance of the in-house vs commercial kit-based approach. Finally, we propose an algorithm aimed at integrating the present strategies of the laboratory diagnostics in AE for the best clinical management of patients with these disorders.
Collapse
Affiliation(s)
- Stefano Masciocchi
- Neuroimmunology Research Section, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, Università degli Studi di Pavia, Pavia, Italy
| | - Pietro Businaro
- Neuroimmunology Research Section, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, Università degli Studi di Pavia, Pavia, Italy
| | - Silvia Scaranzin
- Neuroimmunology Research Section, IRCCS Mondino Foundation, Pavia, Italy
| | - Chiara Morandi
- Neuroimmunology Research Section, IRCCS Mondino Foundation, Pavia, Italy
| | - Diego Franciotta
- Neuroimmunology Research Section, IRCCS Mondino Foundation, Pavia, Italy
| | - Matteo Gastaldi
- Neuroimmunology Research Section, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
48
|
Chen M, Koopmans F, Gonzalez-Lozano MA, Smit AB, Li KW. Brain Region Differences in α1- and α5-Subunit-Containing GABA A Receptor Proteomes Revealed with Affinity Purification and Blue Native PAGE Proteomics. Cells 2023; 13:14. [PMID: 38201218 PMCID: PMC10778189 DOI: 10.3390/cells13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
GABAA receptors are the major inhibitory receptors in the brain. They are hetero-pentamers with a composition of predominantly two α, two β, and one γ or δ subunit. Of the six α subunit genes, the α5 subunit displays a limited spatial expression pattern and is known to mediate both phasic and tonic inhibition. In this study, using immunoaffinity-based proteomics, we identified the α5 subunit containing receptor complexes in the hippocampus and olfactory bulb. The α1-α5 interaction was identified in both brain regions, albeit with significantly different stoichiometries. In line with this, reverse IPs using anti-α1 antibodies showed the α5-α1 co-occurrence and validated the quantitative difference. In addition, we showed that the association of Neuroligin 2 with α1-containing receptors was much higher in the olfactory bulb than in the hippocampus, which was confirmed using blue native gel electrophoresis and quantitative mass spectrometry. Finally, immunocytochemical staining revealed a co-localization of α1 and α5 subunits in the post-synaptic puncta in the hippocampus.
Collapse
Affiliation(s)
| | | | | | | | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (M.C.); (M.A.G.-L.); (A.B.S.)
| |
Collapse
|
49
|
Kasaragod VB, Malinauskas T, Wahid AA, Lengyel J, Knoflach F, Hardwick SW, Jones CF, Chen WN, Lucas X, El Omari K, Chirgadze DY, Aricescu AR, Cecere G, Hernandez MC, Miller PS. The molecular basis of drug selectivity for α5 subunit-containing GABA A receptors. Nat Struct Mol Biol 2023; 30:1936-1946. [PMID: 37903907 PMCID: PMC10716045 DOI: 10.1038/s41594-023-01133-1] [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: 02/28/2023] [Accepted: 09/20/2023] [Indexed: 11/01/2023]
Abstract
α5 subunit-containing γ-aminobutyric acid type A (GABAA) receptors represent a promising drug target for neurological and neuropsychiatric disorders. Altered expression and function contributes to neurodevelopmental disorders such as Dup15q and Angelman syndromes, developmental epilepsy and autism. Effective drug action without side effects is dependent on both α5-subtype selectivity and the strength of the positive or negative allosteric modulation (PAM or NAM). Here we solve structures of drugs bound to the α5 subunit. These define the molecular basis of binding and α5 selectivity of the β-carboline, methyl 6,7-dimethoxy-4-ethyl-β-carboline-3-carboxylate (DMCM), type II benzodiazepine NAMs, and a series of isoxazole NAMs and PAMs. For the isoxazole series, each molecule appears as an 'upper' and 'lower' moiety in the pocket. Structural data and radioligand binding data reveal a positional displacement of the upper moiety containing the isoxazole between the NAMs and PAMs. Using a hybrid molecule we directly measure the functional contribution of the upper moiety to NAM versus PAM activity. Overall, these structures provide a framework by which to understand distinct modulator binding modes and their basis of α5-subtype selectivity, appreciate structure-activity relationships, and empower future structure-based drug design campaigns.
Collapse
Affiliation(s)
- Vikram Babu Kasaragod
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ayla A Wahid
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Judith Lengyel
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Frederic Knoflach
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Steven W Hardwick
- CryoEM Facility, Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Wan-Na Chen
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Xavier Lucas
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland
| | - Kamel El Omari
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - Dimitri Y Chirgadze
- CryoEM Facility, Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - A Radu Aricescu
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Giuseppe Cecere
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland
| | - Maria-Clemencia Hernandez
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland.
| | - Paul S Miller
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
50
|
Bhave K, Forman SA. Substituted Cysteine Modification and Protection with n-Alkyl-MTS Reagents Quantifies Steric Changes Induced by a Mutation in Anesthetic Binding Sites on GABA Type A Receptors. Mol Pharmacol 2023; 104:266-274. [PMID: 37586749 PMCID: PMC10658906 DOI: 10.1124/molpharm.123.000719] [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: 05/08/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/18/2023] Open
Abstract
Multiple approaches, including cryogenic electron microscopy (cryo-EM), indicate that the anesthetics etomidate and propofol modulate α1β2/3γ2 GABAA receptors by binding in overlapping transmembrane inter-subunit sites near βM286 and αL232 sidechains. High-precision approaches in functional receptors are needed for comparisons with cryo-EM. We previously used substituted cysteine modification and protection (SCAMP) with n-alkyl-methanethiosulfonate (MTS) reagents and electrophysiology in α1β3M286Cγ2L receptors to estimate the distance from etomidate to β3M286 with precision near 1.3 Å. Here, we address three more aims using this approach: (i) SCAMP with etomidate was tested in α1L232Cβ3γ2L receptors; (ii) studies in α1L232Wβ3M286Cγ2L receptors assessed whether α1L232W displaces etomidate relative to β3M286C; and (iii) results with propofol were compared with those with etomidate. Voltage-clamp electrophysiology in Xenopus oocytes was used to assess persistent functional changes after exposing cysteine-substituted receptors to methyl-MTS through n-decyl-MTS. Overlap of modified cysteine sidechains with bound anesthetic was inferred when anesthetic co-application with alkyl-MTS reagent blocked the development of persistent effects. In α1L232Cβ3γ2L receptors, only pentyl-MTS and hexyl-MTS induced persistent effects that were unaltered by etomidate co-application, precluding a direct estimate of intermolecular distance. In α1L232Wβ3M286Cγ2L receptors, sidechain overlap with bound etomidate was inferred for modifications with ethyl-MTS through n-pentyl-MTS, with unambiguous cut-on and cut-off. Comparison with results in α1β3M286Cγ2L reveals that α1L232W, which increases maximal sidechain length by 2.1 Å, displaces etomidate closer to β3M286C by about 1.3 Å. Propofol results largely mirrored those with etomidate. These findings indicate that both etomidate and propofol bind within 1 Å of α1L232, consistent with cryo-EM structures. SIGNIFICANCE STATEMENT: We combined electrophysiology, cysteine substitutions, and n-alkyl-methanethiosulfonate modifiers in functional GABAA receptors to enable precise estimates of the distance between β3M286C sidechains and anesthetics (etomidate and propofol) bound in transmembrane β+/α- inter-subunit pockets. Comparing results in α1β3M286Cγ2L and α1L232Wβ3M286Cγ2L receptors reveals that α1L232W mutations displace both anesthetics toward β3M286C, indicating that these anesthetics bind within 1 Å of the α1L232 sidechain in functional receptors, consistent with cryogenic electron microscopy structures derived under nonphysiologic conditions.
Collapse
Affiliation(s)
- Kieran Bhave
- Beecher-Mallinckrodt Laboratories, Department of Anesthesia Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Stuart A Forman
- Beecher-Mallinckrodt Laboratories, Department of Anesthesia Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|