1
|
Peterson L, Nguyen J, Ghani N, Rodriguez-Echemendia P, Qiao H, Guwn SY, Man HY, Kantak KM. Molecular mechanisms underlying sex and treatment-dependent differences in an animal model of cue-exposure therapy for cocaine relapse prevention. Front Neurosci 2024; 18:1425447. [PMID: 39176383 PMCID: PMC11339646 DOI: 10.3389/fnins.2024.1425447] [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: 04/29/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Environmental enrichment combined with the glycine transporter-1 inhibitor Org24598 (EE+ORG) during cocaine-cue extinction (EXT) inhibited reacquisition of 1.0 mg/kg cocaine self-administration in male but not female rats in a previous investigation. In this investigation, we determined if this treatment benefit in males required EXT training and ascertained the molecular basis for the observed sex difference in treatment efficacy. Nine groups of male rats trained to self-administer 1.0 mg/kg cocaine or receiving yoked-saline underwent EXT or NoEXT with or without EE and/or ORG. Next, they underwent reacquisition of cocaine self-administration or were sacrificed for molecular analysis of 9 protein targets indicative of neuroplasticity in four brain regions. Two groups of female rats trained to self-administer 1.0 mg/kg cocaine also underwent EXT with or without EE + ORG and were sacrificed for molecular analysis, as above. EE + ORG facilitated the rate of EXT learning in both sexes, and importantly, the therapeutic benefit of EE + ORG for inhibiting cocaine relapse required EXT training. Males were more sensitive than females to neuroplasticity-inducing effects of EE + ORG, which prevented reductions in total GluA1 and PSD95 proteins selectively in basolateral amygdala of male rats trained to self-administer cocaine and receiving EXT. Females were deficient in expression of multiple protein targets, especially after EE + ORG. These included total GluA1 and PSD95 proteins in basolateral amygdala, and total TrkB protein in basolateral amygdala, dorsal hippocampus, and ventromedial prefrontal cortex. Together, these results support the clinical view that sex-specific pharmacological and behavioral treatment approaches may be needed during cue exposure therapy to inhibit cocaine relapse.
Collapse
Affiliation(s)
- Lucy Peterson
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biology, Boston University, Boston, MA, United States
| | - Jonathan Nguyen
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, United States
| | - Naveed Ghani
- Department of Biology, Boston University, Boston, MA, United States
| | | | - Hui Qiao
- Department of Biology, Boston University, Boston, MA, United States
| | - Sun Young Guwn
- Department of Biology, Boston University, Boston, MA, United States
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, United States
| | - Kathleen M. Kantak
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, United States
| |
Collapse
|
2
|
Supplisson S. Dynamic role of GlyT1 as glycine sink or source: Pharmacological implications for the gain control of NMDA receptors. Neuroscience 2024:S0306-4522(24)00350-6. [PMID: 39059742 DOI: 10.1016/j.neuroscience.2024.07.037] [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: 02/27/2024] [Revised: 07/03/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Glycine transporter 1 (GlyT1) mediates the termination of inhibitory glycinergic receptor signaling in the spinal cord and brainstem, and is also present diffusely in the forebrain. Here, it regulates the ambient glycine concentration and influences the 'glycine' site occupancy of N-methyl-d-aspartate receptors (NMDARs). GlyT1 is a reversible transporter with a substantial, but not excessive, sodium-motive force for uphill transport. This study investigates its role as a potential source of glycine supply, either by reverse uptake or heteroexchange. Indeed, glutamate alone does not induce NMDAR current in "naive" oocytes co-expressing GluN1/GluN2A and GlyT1, a previously characterized cellular model. However, after substantial intracellular glycine accumulation, GlyT1 reverses its transport mode, and begins to release glycine into the external compartment, allowing NMDAR activation by glutamate alone. These uptake-dependent glutamate currents were blocked by ALX-5407 and potentiated by sarcosine, a specific inhibitor and substrate of GlyT1, respectively, suggesting a higher occupancy of the co-agonist site when GlyT1 functions as a glycine source either by reversed-uptake or by heteroexchange. These two glycine release mechanisms can be distinguished by their voltage dependence, as the reversed-uptake cycle decreases at hyperpolarized potentials, whereas heteroexchange electroneutrality preserves glycine efflux and NMDAR activation at these potentials. These results establish GlyT1-mediated efflux as a positive regulator of NMDAR coagonist site occupancy, and demonstrate the efficacy of sarcosine heteroexchange in enhancing coagonist site occupancy. Because NMDAR facilitation by GlyT1-inhibitors and sarcosine relies on different transport mechanisms, their actions may be a source of variability in reversing NMDAR hypofunction in schizophrenia.
Collapse
Affiliation(s)
- Stéphane Supplisson
- Institut de Biologie de l'ENS (IBENS), Ecole normale supérieure, Université PSL, CNRS, INSERM, Paris, F-75005, France.
| |
Collapse
|
3
|
Seljeset S, Sintsova O, Wang Y, Harb HY, Lynagh T. Constitutive activity of ionotropic glutamate receptors via hydrophobic substitutions in the ligand-binding domain. Structure 2024; 32:966-978.e6. [PMID: 38677289 DOI: 10.1016/j.str.2024.04.001] [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: 08/11/2023] [Revised: 02/11/2024] [Accepted: 04/02/2024] [Indexed: 04/29/2024]
Abstract
Neurotransmitter ligands electrically excite neurons by activating ionotropic glutamate receptor (iGluR) ion channels. Knowledge of the iGluR amino acid residues that dominate ligand-induced activation would enable the prediction of function from sequence. We therefore explored the molecular determinants of activity in rat N-methyl-D-aspartate (NMDA)-type iGluRs (NMDA receptors), complex heteromeric iGluRs comprising two glycine-binding GluN1 and two glutamate-binding GluN2 subunits, using amino acid sequence analysis, mutagenesis, and electrophysiology. We find that a broadly conserved aspartate residue controls both ligand potency and channel activity, to the extent that certain substitutions at this position bypass the need for ligand binding in GluN1 subunits, generating NMDA receptors activated solely by glutamate. Furthermore, we identify a homomeric iGluR from the placozoan Trichoplax adhaerens that has utilized native mutations of this crucial residue to evolve into a leak channel that is inhibited by neurotransmitter binding, pointing to a dominant role of this residue throughout the iGluR superfamily.
Collapse
Affiliation(s)
- Sandra Seljeset
- Michael Sars Centre, University of Bergen, 5008 Bergen, Norway
| | - Oksana Sintsova
- Michael Sars Centre, University of Bergen, 5008 Bergen, Norway
| | - Yuhong Wang
- Michael Sars Centre, University of Bergen, 5008 Bergen, Norway
| | - Hassan Y Harb
- Concept Life Sciences Limited, Frith Knoll Road, Chapel-en-le-Frith, SK23 0PG High Peak, UK
| | - Timothy Lynagh
- Michael Sars Centre, University of Bergen, 5008 Bergen, Norway.
| |
Collapse
|
4
|
Korinek M, Candelas Serra M, Abdel Rahman F, Dobrovolski M, Kuchtiak V, Abramova V, Fili K, Tomovic E, Hrcka Krausova B, Krusek J, Cerny J, Vyklicky L, Balik A, Smejkalova T. Disease-Associated Variants in GRIN1, GRIN2A and GRIN2B genes: Insights into NMDA Receptor Structure, Function, and Pathophysiology. Physiol Res 2024; 73:S413-S434. [PMID: 38836461 PMCID: PMC11412357 DOI: 10.33549/physiolres.935346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are a subtype of ionotropic glutamate receptors critical for synaptic transmission and plasticity, and for the development of neural circuits. Rare or de-novo variants in GRIN genes encoding NMDAR subunits have been associated with neurodevelopmental disorders characterized by intellectual disability, developmental delay, autism, schizophrenia, or epilepsy. In recent years, some disease-associated variants in GRIN genes have been characterized using recombinant receptors expressed in non-neuronal cells, and a few variants have also been studied in neuronal preparations or animal models. Here we review the current literature on the functional evaluation of human disease-associated variants in GRIN1, GRIN2A and GRIN2B genes at all levels of analysis. Focusing on the impact of different patient variants at the level of receptor function, we discuss effects on receptor agonist and co-agonist affinity, channel open probability, and receptor cell surface expression. We consider how such receptor-level functional information may be used to classify variants as gain-of-function or loss-of-function, and discuss the limitations of this classification at the synaptic, cellular, or system level. Together this work by many laboratories worldwide yields valuable insights into NMDAR structure and function, and represents significant progress in the effort to understand and treat GRIN disorders. Keywords: NMDA receptor , GRIN genes, Genetic variants, Electrophysiology, Synapse, Animal models.
Collapse
Affiliation(s)
- M Korinek
- Department of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Izidoro Ribeiro R, Almeida Carvalho G, Almeida Chiareli R, Vieira de Assis Lima I, Quaglio Bellozi PM, Oliveira-Lima OC, Oliveira Giacomelli Á, Birbrair A, Santiago Gomez R, Pinheiro de Oliveira AC, Ulrich H, Cunha Xavier Pinto M. Glycine transporter-1 inhibition by NFPS promotes neuroprotection against striatal damage models. Neurosci Lett 2024; 826:137715. [PMID: 38460902 DOI: 10.1016/j.neulet.2024.137715] [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: 12/24/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
The striatum, an essential component of the brain's motor and reward systems, plays a pivotal role in a wide array of cognitive processes. Its dysfunction is a hallmark of neurodegenerative diseases like Parkinson's disease (PD) and Huntington's disease (HD), leading to profound motor and cognitive deficits. These conditions are often related to excitotoxicity, primarily due to overactivation of NMDA receptors (NMDAR). In the synaptic cleft, glycine transporter type 1 (GlyT1) controls the glycine levels, a NMDAR co-agonist, which modulates NMDAR function. This research explored the neuroprotective potential of NFPS, a GlyT1 inhibitor, in murine models of striatal injury. Employing models of neurotoxicity induced by 6-hydroxydopamine (PD model) and quinolinic acid (HD model), we assessed the effectiveness of NFPS pre-treatment in maintaining the integrity of striatal neurons and averting neuronal degeneration. The results indicated that NFPS pre-treatment conferred significant neuroprotection, reducing neuronal degeneration, protecting dopaminergic neurons, and preserving dendritic spines within the striatum. Additionally, this pre-treatment notably mitigated motor impairments resulting from striatal damage. The study revealed that GlyT1 inhibition led to substantial changes in the ratios of NMDAR subunits GluN2A/GluN1 and GluN2B/GluN1, 24 h after NFPS treatment. These findings underscore the neuroprotective efficacy of GlyT1 inhibition, proposing it as a viable therapeutic strategy for striatum-related damage.
Collapse
Affiliation(s)
- Raul Izidoro Ribeiro
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Gustavo Almeida Carvalho
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Raphaela Almeida Chiareli
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Isabel Vieira de Assis Lima
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | - Paula Maria Quaglio Bellozi
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | | | | | - Alexander Birbrair
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | - Renato Santiago Gomez
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | | | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo-SP, Brazil
| | - Mauro Cunha Xavier Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil.
| |
Collapse
|
6
|
Moss A, Kuttippurathu L, Srivastava A, Schwaber JS, Vadigepalli R. Dynamic dysregulation of transcriptomic networks in brainstem autonomic nuclei during hypertension development in the female spontaneously hypertensive rat. Physiol Genomics 2024; 56:283-300. [PMID: 38145287 PMCID: PMC11283910 DOI: 10.1152/physiolgenomics.00073.2023] [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: 07/21/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
Neurogenic hypertension stems from an imbalance in autonomic function that shifts the central cardiovascular control circuits toward a state of dysfunction. Using the female spontaneously hypertensive rat and the normotensive Wistar-Kyoto rat model, we compared the transcriptomic changes in three autonomic nuclei in the brainstem, nucleus of the solitary tract (NTS), caudal ventrolateral medulla, and rostral ventrolateral medulla (RVLM) in a time series at 8, 10, 12, 16, and 24 wk of age, spanning the prehypertensive stage through extended chronic hypertension. RNA-sequencing data were analyzed using an unbiased, dynamic pattern-based approach that uncovered dominant and several subtle differential gene regulatory signatures. Our results showed a persistent dysregulation across all three autonomic nuclei regardless of the stage of hypertension development as well as a cascade of transient dysregulation beginning in the RVLM at the prehypertensive stage that shifts toward the NTS at the hypertension onset. Genes that were persistently dysregulated were heavily enriched for immunological processes such as antigen processing and presentation, the adaptive immune response, and the complement system. Genes with transient dysregulation were also largely region-specific and were annotated for processes that influence neuronal excitability such as synaptic vesicle release, neurotransmitter transport, and an array of neuropeptides and ion channels. Our results demonstrate that neurogenic hypertension is characterized by brainstem region-specific transcriptomic changes that are highly dynamic with significant gene regulatory changes occurring at the hypertension onset as a key time window for dysregulation of homeostatic processes across the autonomic control circuits.NEW & NOTEWORTHY Hypertension is a major disease and is the primary risk factor for cardiovascular complications and stroke. The gene expression changes in the central nervous system circuits driving hypertension are understudied. Here, we show that coordinated and region-specific gene expression changes occur in the brainstem autonomic circuits over time during the development of a high blood pressure phenotype in a rat model of human essential hypertension.
Collapse
Affiliation(s)
- Alison Moss
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Lakshmi Kuttippurathu
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Ankita Srivastava
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - James S Schwaber
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
7
|
Cortese K, Gagliani MC, Raiteri L. Interactions between Glycine and Glutamate through Activation of Their Transporters in Hippocampal Nerve Terminals. Biomedicines 2023; 11:3152. [PMID: 38137373 PMCID: PMC10740625 DOI: 10.3390/biomedicines11123152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Evidence supports the pathophysiological relevance of crosstalk between the neurotransmitters Glycine and Glutamate and their close interactions; some reports even support the possibility of Glycine-Glutamate cotransmission in central nervous system (CNS) areas, including the hippocampus. Functional studies with isolated nerve terminals (synaptosomes) permit us to study transporter-mediated interactions between neurotransmitters that lead to the regulation of transmitter release. Our main aims here were: (i) to investigate release-regulating, transporter-mediated interactions between Glycine and Glutamate in hippocampal nerve terminals and (ii) to determine the coexistence of transporters for Glycine and Glutamate in these terminals. Purified synaptosomes, analyzed at the ultrastructural level via electron microscopy, were used as the experimental model. Mouse hippocampal synaptosomes were prelabeled with [3H]D-Aspartate or [3H]Glycine; the release of radiolabeled tracers was monitored with the superfusion technique. The main findings were that (i) exogenous Glycine stimulated [3H]D-Aspartate release, partly by activation of GlyT1 and in part, unusually, through GlyT2 transporters and that (ii) D-Aspartate stimulated [3H]glycine release by a process that was sensitive to Glutamate transporter blockers. Based on the features of the experimental model used, it is suggested that functional transporters for Glutamate and Glycine coexist in a small subset of hippocampal nerve terminals, a condition that may also be compatible with cotransmission; glycinergic and glutamatergic transporters exhibit different functions and mediate interactions between the neurotransmitters. It is hoped that increased information on Glutamate-Glycine interactions in different areas, including the hippocampus, will contribute to a better knowledge of drugs acting at "glycinergic" targets, currently under study in relation with different CNS pathologies.
Collapse
Affiliation(s)
- Katia Cortese
- Department of Experimental Medicine (DIMES), Cellular Electron Microscopy Lab, University of Genoa, 16132 Genoa, Italy; (K.C.); (M.C.G.)
| | - Maria Cristina Gagliani
- Department of Experimental Medicine (DIMES), Cellular Electron Microscopy Lab, University of Genoa, 16132 Genoa, Italy; (K.C.); (M.C.G.)
| | - Luca Raiteri
- Department of Pharmacy (DIFAR), Pharmacology and Toxicology Section, University of Genoa, 16148 Genoa, Italy
| |
Collapse
|
8
|
Latli B, Hrapchak MJ, Frutos RP, Lee H, Song JJ. Carbon 14 synthesis of glycine transporter 1 inhibitor Iclepertin (BI 425809) and its major metabolites. J Labelled Comp Radiopharm 2023; 66:336-344. [PMID: 37382087 DOI: 10.1002/jlcr.4051] [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: 05/07/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
Carbon 14 labeled Iclepertin (BI 425809, 1) and its major metabolites were needed for ADME and several other studies necessary for the advancement of this drug candidate in clinical trials. Iclepertin is composed of two main chemical blocks, (R)-5-(methylsulfonyl)-2-([1,1,1-trifluoropropan-2-yl]oxy)benzoic acid (2), and 3-[(1R,5R)-3-azabicyclo[3.1.0]hexan-5-yl]-5-(trifluoromethyl)isoxazole (3) linked to each other via an amide bond. In the first synthesis of carbon 14 labeled 1, 2-fluorobenzoic acid, carboxyl-14 C was converted to [14 C]-2 in three steps and then coupled to 3 to provide [14 C]-1a in 45% overall yield. In the second synthesis, [14 C]-3 was prepared in six radioactive steps and coupled to the acid 2 to furnish [14 C]-1b in 20% overall yield. Both synthetic routes provided [14 C]-1a and [14 C]-1b with specific activities higher than 53 mCi/mmol and radiochemical, chemical, and enantiomeric purities above 98%. Two major metabolites of 1, BI 761036 and BI 758790, were also prepared labeled with carbon 14 using intermediates already available from the synthesis of [14 C]-1.
Collapse
Affiliation(s)
- Bachir Latli
- The Radiosynthesis Laboratory, Chemical Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Matt J Hrapchak
- The Radiosynthesis Laboratory, Chemical Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Rogelio P Frutos
- The Radiosynthesis Laboratory, Chemical Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Heewon Lee
- The Radiosynthesis Laboratory, Chemical Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Jinhua J Song
- The Radiosynthesis Laboratory, Chemical Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| |
Collapse
|
9
|
Aguayo-Cerón KA, Sánchez-Muñoz F, Gutierrez-Rojas RA, Acevedo-Villavicencio LN, Flores-Zarate AV, Huang F, Giacoman-Martinez A, Villafaña S, Romero-Nava R. Glycine: The Smallest Anti-Inflammatory Micronutrient. Int J Mol Sci 2023; 24:11236. [PMID: 37510995 PMCID: PMC10379184 DOI: 10.3390/ijms241411236] [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: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Glycine is a non-essential amino acid with many functions and effects. Glycine can bind to specific receptors and transporters that are expressed in many types of cells throughout an organism to exert its effects. There have been many studies focused on the anti-inflammatory effects of glycine, including its abilities to decrease pro-inflammatory cytokines and the concentration of free fatty acids, to improve the insulin response, and to mediate other changes. However, the mechanism through which glycine acts is not clear. In this review, we emphasize that glycine exerts its anti-inflammatory effects throughout the modulation of the expression of nuclear factor kappa B (NF-κB) in many cells. Although glycine is a non-essential amino acid, we highlight how dietary glycine supplementation is important in avoiding the development of chronic inflammation.
Collapse
Affiliation(s)
- Karla Aidee Aguayo-Cerón
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología "Ignacio Chávez", Ciudad de Mexico 14080, Mexico
| | | | | | - Aurora Vanessa Flores-Zarate
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Fengyang Huang
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de Mexico 06720, Mexico
| | - Abraham Giacoman-Martinez
- Laboratorio de Framacología, Departamaneto de Ciencias de la Salud, DCBS, Universidad Autónoma Mteropolitana-Iztapalapa (UAM-I), Ciudad de Mexico 09340, Mexico
| | - Santiago Villafaña
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Rodrigo Romero-Nava
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| |
Collapse
|
10
|
Surti TS, Ranganathan M, Johannesen JK, Gueorguieva R, Deaso E, Kenney JG, Krystal JH, D'Souza DC. Randomized controlled trial of the glycine transporter 1 inhibitor PF-03463275 to enhance cognitive training and neuroplasticity in schizophrenia. Schizophr Res 2023; 256:36-43. [PMID: 37141764 PMCID: PMC10257994 DOI: 10.1016/j.schres.2023.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 03/23/2023] [Accepted: 04/19/2023] [Indexed: 05/06/2023]
Abstract
N-methyl-d-aspartate glutamate receptor (NMDAR) hypofunction is implicated in the impaired neuroplasticity and cognitive impairments associated with schizophrenia (CIAS). We hypothesized that enhancing NMDAR function by inhibiting the glycine transporter-1 (GLYT1) would improve neuroplasticity and thereby augment benefits of non-pharmacological cognitive training (CT) strategies. This study examined whether co-administration of a GLYT1 inhibitor and computerized CT would have synergistic effects on CIAS. Stable outpatients with schizophrenia participated in this double-blind, placebo-controlled, within-subject, crossover augmentation study. Participants received placebo or GLYT1 inhibitor (PF-03463275) for two 5-week periods separated by 2 weeks of washout. PF-03463275 doses (40 or 60 mg twice daily) were selected to produce high GLYT1 occupancy. To limit pharmacodynamic variability, only cytochrome P450 2D6 extensive metabolizers were included. Medication adherence was confirmed daily. Participants received 4 weeks of CT in each treatment period. Cognitive performance (MATRICS Consensus Cognitive Battery) and psychotic symptoms (Positive and Negative Syndrome Scale) were assessed in each period. 71 participants were randomized. PF-03463275 in combination with CT was feasible, safe, and well-tolerated at the doses prescribed but did not produce greater improvement in CIAS compared to CT alone. PF-03463275 was not associated with improved CT learning parameters. Participation in CT was associated with improvement in MCCB scores.
Collapse
Affiliation(s)
- Toral S Surti
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, United States of America; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, United States of America; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States of America.
| | - Mohini Ranganathan
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, United States of America; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, United States of America; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States of America
| | - Jason K Johannesen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States of America
| | - Ralitza Gueorguieva
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States of America
| | - Emma Deaso
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, United States of America; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, United States of America; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States of America
| | - Joshua G Kenney
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States of America
| | - John H Krystal
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, United States of America; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, United States of America; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States of America; Mental Health Service Line, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States of America
| | - Deepak Cyril D'Souza
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, United States of America; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, United States of America; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States of America
| |
Collapse
|
11
|
Piniella D, Zafra F. Functional crosstalk of the glycine transporter GlyT1 and NMDA receptors. Neuropharmacology 2023; 232:109514. [PMID: 37003571 DOI: 10.1016/j.neuropharm.2023.109514] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
NMDA-type glutamate receptors (NMDARs) constitute one of the main glutamate (Glu) targets in the central nervous system and are involved in synaptic plasticity, which is the molecular substrate of learning and memory. Hypofunction of NMDARs has been associated with schizophrenia, while overstimulation causes neuronal death in neurodegenerative diseases or in stroke. The function of NMDARs requires coincidental binding of Glu along with other cellular signals such as neuronal depolarization, and the presence of other endogenous ligands that modulate their activity by allosterism. Among these allosteric modulators are zinc, protons and Gly, which is an obligatory co-agonist. These characteristics differentiate NMDARs from other receptors, and their structural bases have begun to be established in recent years. In this review we focus on the crosstalk between Glu and glycine (Gly), whose concentration in the NMDAR microenvironment is maintained by various Gly transporters that remove or release it into the medium in a regulated manner. The GlyT1 transporter is particularly involved in this task, and has become a target of great interest for the treatment of schizophrenia since its inhibition leads to an increase in synaptic Gly levels that enhances the activity of NMDARs. However, the only drug that has completed phase III clinical trials did not yield the expected results. Notwithstanding, there are additional drugs that continue to be investigated, and it is hoped that knowledge gained from the recently published 3D structure of GlyT1 may allow the rational design of more effective new drugs.
Collapse
Affiliation(s)
- Dolores Piniella
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain; IdiPAZ, Institute of Health Carlos III (ISCIII), Spain
| | - Francisco Zafra
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain; IdiPAZ, Institute of Health Carlos III (ISCIII), Spain.
| |
Collapse
|
12
|
An astrocytic signaling loop for frequency-dependent control of dendritic integration and spatial learning. Nat Commun 2022; 13:7932. [PMID: 36566254 PMCID: PMC9789958 DOI: 10.1038/s41467-022-35620-8] [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: 01/04/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022] Open
Abstract
Dendrites of hippocampal CA1 pyramidal cells amplify clustered glutamatergic input by activation of voltage-gated sodium channels and N-methyl-D-aspartate receptors (NMDARs). NMDAR activity depends on the presence of NMDAR co-agonists such as D-serine, but how co-agonists influence dendritic integration is not well understood. Using combinations of whole-cell patch clamp, iontophoretic glutamate application, two-photon excitation fluorescence microscopy and glutamate uncaging in acute rat and mouse brain slices we found that exogenous D-serine reduced the threshold of dendritic spikes and increased their amplitude. Triggering an astrocytic mechanism controlling endogenous D-serine supply via endocannabinoid receptors (CBRs) also increased dendritic spiking. Unexpectedly, this pathway was activated by pyramidal cell activity primarily in the theta range, which required HCN channels and astrocytic CB1Rs. Therefore, astrocytes close a positive and frequency-dependent feedback loop between pyramidal cell activity and their integration of dendritic input. Its disruption in mice led to an impairment of spatial memory, which demonstrated its behavioral relevance.
Collapse
|
13
|
Kuo CY, Lin CH, Lane HY. Targeting D-Amino Acid Oxidase (DAAO) for the Treatment of Schizophrenia: Rationale and Current Status of Research. CNS Drugs 2022; 36:1143-1153. [PMID: 36194364 DOI: 10.1007/s40263-022-00959-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2022] [Indexed: 11/29/2022]
Abstract
In the brain, D-amino acid oxidase (DAAO) is a peroxisomal flavoenzyme. Through oxidative deamination by DAAO, D-serine, the main coagonist of synaptic N-methyl-D-aspartate receptors (NMDARs), is degraded into α-keto acids and ammonia; flavin adenine dinucleotide (FAD) is simultaneously reduced to dihydroflavine-adenine dinucleotide (FADH2), which is subsequently reoxidized to FAD, with hydrogen peroxide produced as a byproduct. NMDAR hypofunction is implicated in the pathogenesis of schizophrenia. In previous studies, compared with control subjects, patients with schizophrenia had lower D-serine levels in peripheral blood and cerebrospinal fluid but higher DAAO expression and activity in the brain. Inhibiting DAAO activity and slowing D-serine degradation by using DAAO inhibitors to enhance NMDAR function may be a new strategy for use in the treatment of schizophrenia. The aim of this leading article is to review the current research in DAAO inhibitors.
Collapse
Affiliation(s)
- Chien-Yi Kuo
- Department of Psychiatry, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung City, 404327, Taiwan, ROC.,Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Rd., North Dis., Taichung City, 404333, Taiwan, ROC
| | - Chieh-Hsin Lin
- Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Rd., North Dis., Taichung City, 404333, Taiwan, ROC. .,Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, No. 123, Dapi Rd., Niaosong Dist., Kaohsiung City, 83301, Taiwan, ROC. .,School of Medicine, Chang Gung University, No. 259, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 33302, Taiwan, ROC.
| | - Hsien-Yuan Lane
- Department of Psychiatry, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung City, 404327, Taiwan, ROC. .,Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Rd., North Dis., Taichung City, 404333, Taiwan, ROC. .,Department of Psychology, College of Medical and Health Sciences, Asia University, No. 500, Lioufeng Rd., Wufeng Dist., Taichung City, 413305, Taiwan, ROC.
| |
Collapse
|
14
|
Sivakumar S, Ghasemi M, Schachter SC. Targeting NMDA Receptor Complex in Management of Epilepsy. Pharmaceuticals (Basel) 2022; 15:ph15101297. [PMID: 36297409 PMCID: PMC9609646 DOI: 10.3390/ph15101297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are widely distributed in the central nervous system (CNS) and play critical roles in neuronal excitability in the CNS. Both clinical and preclinical studies have revealed that the abnormal expression or function of these receptors can underlie the pathophysiology of seizure disorders and epilepsy. Accordingly, NMDAR modulators have been shown to exert anticonvulsive effects in various preclinical models of seizures, as well as in patients with epilepsy. In this review, we provide an update on the pathologic role of NMDARs in epilepsy and an overview of the NMDAR antagonists that have been evaluated as anticonvulsive agents in clinical studies, as well as in preclinical seizure models.
Collapse
Affiliation(s)
- Shravan Sivakumar
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
- Correspondence: (M.G.); (S.C.S.)
| | - Steven C. Schachter
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02114, USA
- Consortia for Improving Medicine with Innovation & Technology (CIMIT), Boston, MA 02114, USA
- Correspondence: (M.G.); (S.C.S.)
| |
Collapse
|
15
|
de Bartolomeis A, Vellucci L, Austin MC, De Simone G, Barone A. Rational and Translational Implications of D-Amino Acids for Treatment-Resistant Schizophrenia: From Neurobiology to the Clinics. Biomolecules 2022; 12:biom12070909. [PMID: 35883465 PMCID: PMC9312470 DOI: 10.3390/biom12070909] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia has been conceptualized as a neurodevelopmental disorder with synaptic alterations and aberrant cortical–subcortical connections. Antipsychotics are the mainstay of schizophrenia treatment and nearly all share the common feature of dopamine D2 receptor occupancy, whereas glutamatergic abnormalities are not targeted by the presently available therapies. D-amino acids, acting as N-methyl-D-aspartate receptor (NMDAR) modulators, have emerged in the last few years as a potential augmentation strategy in those cases of schizophrenia that do not respond well to antipsychotics, a condition defined as treatment-resistant schizophrenia (TRS), affecting almost 30–40% of patients, and characterized by serious cognitive deficits and functional impairment. In the present systematic review, we address with a direct and reverse translational perspective the efficacy of D-amino acids, including D-serine, D-aspartate, and D-alanine, in poor responders. The impact of these molecules on the synaptic architecture is also considered in the light of dendritic spine changes reported in schizophrenia and antipsychotics’ effect on postsynaptic density proteins. Moreover, we describe compounds targeting D-amino acid oxidase and D-aspartate oxidase enzymes. Finally, other drugs acting at NMDAR and proxy of D-amino acids function, such as D-cycloserine, sarcosine, and glycine, are considered in the light of the clinical burden of TRS, together with other emerging molecules.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
- Correspondence: ; Tel.: +39-081-7463673 or +39-081-7463884 or +39-3662745592; Fax: +39-081-7462644
| | - Licia Vellucci
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| | - Mark C. Austin
- Clinical Psychopharmacology Program, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA;
| | - Giuseppe De Simone
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| | - Annarita Barone
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| |
Collapse
|
16
|
Peiser-Oliver JM, Evans S, Adams DJ, Christie MJ, Vandenberg RJ, Mohammadi SA. Glycinergic Modulation of Pain in Behavioral Animal Models. Front Pharmacol 2022; 13:860903. [PMID: 35694265 PMCID: PMC9174897 DOI: 10.3389/fphar.2022.860903] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/19/2022] [Indexed: 11/16/2022] Open
Abstract
Animal models of human pain conditions allow for detailed interrogation of known and hypothesized mechanisms of pain physiology in awake, behaving organisms. The importance of the glycinergic system for pain modulation is well known; however, manipulation of this system to treat and alleviate pain has not yet reached the sophistication required for the clinic. Here, we review the current literature on what animal behavioral studies have allowed us to elucidate about glycinergic pain modulation, and the progress toward clinical treatments so far. First, we outline the animal pain models that have been used, such as nerve injury models for neuropathic pain, chemogenic pain models for acute and inflammatory pain, and other models that mimic painful human pathologies such as diabetic neuropathy. We then discuss the genetic approaches to animal models that have identified the crucial glycinergic machinery involved in neuropathic and inflammatory pain. Specifically, two glycine receptor (GlyR) subtypes, GlyRα1(β) and GlyRα3(β), and the two glycine transporters (GlyT), GlyT1 and GlyT2. Finally, we review the different pharmacological approaches to manipulating the glycinergic system for pain management in animal models, such as partial vs. full agonism, reversibility, and multi-target approaches. We discuss the benefits and pitfalls of using animal models in drug development broadly, as well as the progress of glycinergic treatments from preclinical to clinical trials.
Collapse
Affiliation(s)
| | - Sally Evans
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | | | | | - Sarasa A. Mohammadi
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- *Correspondence: Sarasa A. Mohammadi,
| |
Collapse
|
17
|
Nava-Gómez L, Calero-Vargas I, Higinio-Rodríguez F, Vázquez-Prieto B, Olivares-Moreno R, Ortiz-Retana J, Aranda P, Hernández-Chan N, Rojas-Piloni G, Alcauter S, López-Hidalgo M. AGING-ASSOCIATED COGNITIVE DECLINE IS REVERSED BY D-SERINE SUPPLEMENTATION. eNeuro 2022; 9:ENEURO.0176-22.2022. [PMID: 35584913 PMCID: PMC9186414 DOI: 10.1523/eneuro.0176-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 11/21/2022] Open
Abstract
Brain aging is a natural process that involves structural and functional changes that lead to cognitive decline, even in healthy subjects. This detriment has been associated with N-methyl-D-aspartate receptor (NMDAR) hypofunction due to a reduction in the brain levels of D-serine, the endogenous NMDAR co-agonist. However, it is not clear if D-serine supplementation could be used as an intervention to reduce or reverse age-related brain alterations. In the present work, we aimed to analyze the D-serine effect on aging-associated alterations in cellular and large-scale brain systems that could support cognitive flexibility in rats. We found that D-serine supplementation reverts the age-related decline in cognitive flexibility, frontal dendritic spine density, and partially restored large-scale functional connectivity without inducing nephrotoxicity; instead, D-serine restored the thickness of the renal epithelial cells that were affected by age. Our results suggest that D-serine could be used as a therapeutic target to reverse age-related brain alterations.SIGNIFICANT STATEMENTAge-related behavioral changes in cognitive performance occur as a physiological process of aging. Then, it is important to explore possible therapeutics to decrease, retard or reverse aging effects on the brain. NMDA receptor hypofunction contributes to the aging-associated cognitive decline. In the aged brain, there is a reduction in the brain levels of the NMDAR co-agonist, D-Serine. However, it is unclear if chronic D-serine supplementation could revert the age-detriment in brain functions. Our results show that D-serine supplementation reverts the age-associated decrease in cognitive flexibility, functional brain connectivity, and neuronal morphology. Our findings raise the possibility that restoring the brain levels of D-serine could be used as a therapeutic target to recover brain alterations associated with aging.
Collapse
Affiliation(s)
- L Nava-Gómez
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
- Facultad de Medicina. UAQ
| | - I Calero-Vargas
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - F Higinio-Rodríguez
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - B Vázquez-Prieto
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - R Olivares-Moreno
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - J Ortiz-Retana
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - P Aranda
- Facultad de Ciencias Naturales, UAQ
| | | | - G Rojas-Piloni
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - S Alcauter
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - M López-Hidalgo
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
| |
Collapse
|
18
|
Abstract
The brain is a highly energy-demanding organ and requires bioenergetic adaptability to balance normal activity with pathophysiological fuelling of spontaneous recurrent seizures, the hallmark feature of the epilepsies. Recurrent or prolonged seizures have long been known to permanently alter neuronal circuitry and to cause excitotoxic injury and aberrant inflammation. Furthermore, pathological changes in bioenergetics and metabolism are considered downstream consequences of epileptic seizures that begin at the synaptic level. However, as we highlight in this Review, evidence is also emerging that primary derangements in cellular or mitochondrial metabolism can result in seizure genesis and lead to spontaneous recurrent seizures. Basic and translational research indicates that the relationships between brain metabolism and epileptic seizures are complex and bidirectional, producing a vicious cycle that compounds the deleterious consequences of seizures. Metabolism-based treatments such as the high-fat, antiseizure ketogenic diet have become mainstream, and metabolic substrates and enzymes have become attractive molecular targets for seizure prevention and recovery. Moreover, given that metabolism is crucial for epigenetic as well as inflammatory changes, the idea that epileptogenesis can be both negatively and positively influenced by metabolic changes is rapidly gaining ground. Here, we review evidence that supports both pathophysiological and therapeutic roles for brain metabolism in epilepsy.
Collapse
|
19
|
Cappelli J, Khacho P, Wang B, Sokolovski A, Bakkar W, Raymond S, Ahlskog N, Pitney J, Wu J, Chudalayandi P, Wong AYC, Bergeron R. Glycine-induced NMDA receptor internalization provides neuroprotection and preserves vasculature following ischemic stroke. iScience 2022; 25:103539. [PMID: 34977503 PMCID: PMC8689229 DOI: 10.1016/j.isci.2021.103539] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 11/24/2021] [Indexed: 11/26/2022] Open
Abstract
Ischemic stroke is the second leading cause of death worldwide. Following an ischemic event, neuronal death is triggered by uncontrolled glutamate release leading to overactivation of glutamate sensitive N-methyl-d-aspartate receptor (NMDAR). For gating, NMDARs require not only the binding of glutamate, but also of glycine or a glycine-like compound as a co-agonist. Low glycine doses enhance NMDAR function, whereas high doses trigger glycine-induced NMDAR internalization (GINI) in vitro. Here, we report that following an ischemic event, in vivo, GINI also occurs and provides neuroprotection in the presence of a GlyT1 antagonist (GlyT1-A). Mice pretreated with a GlyT1-A, which increases synaptic glycine levels, exhibited smaller stroke volume, reduced cell death, and minimized behavioral deficits following stroke induction by either photothrombosis or endothelin-1. Moreover, we show evidence that in ischemic conditions, GlyT1-As preserve the vasculature in the peri-infarct area. Therefore, GlyT1 could be a new target for the treatment of ischemic stroke. GINI is a dynamic phenomenon which dampens NMDAR-mediated excitotoxicity during stroke GlyT1-antagonists (GlyT1-As) trigger GINI during stroke in vivo GlyT1-As mitigate post-stroke behavioral deficits and preserve peri-infarct vasculature GlyT1 could be a novel and viable therapeutic target for ischemic stroke
Collapse
Affiliation(s)
- Julia Cappelli
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Pamela Khacho
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Boyang Wang
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Alexandra Sokolovski
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Wafae Bakkar
- Ottawa Hospital Research Institute, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Sophie Raymond
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Nina Ahlskog
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Julian Pitney
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Junzheng Wu
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Prakash Chudalayandi
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Adrian Y C Wong
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| | - Richard Bergeron
- Cellular and Molecular Medicine Department, University of Ottawa, 451 Smyth Road, Roger Guindon Building, Room 3501N, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
20
|
Zhao F, Atxabal U, Mariottini S, Yi F, Lotti JS, Rouzbeh N, Liu N, Bunch L, Hansen KB, Clausen RP. Derivatives of ( R)-3-(5-Furanyl)carboxamido-2-aminopropanoic Acid as Potent NMDA Receptor Glycine Site Agonists with GluN2 Subunit-Specific Activity. J Med Chem 2022; 65:734-746. [PMID: 34918931 PMCID: PMC9437973 DOI: 10.1021/acs.jmedchem.1c01810] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
NMDA receptors mediate glutamatergic neurotransmission and are therapeutic targets due to their involvement in a variety of psychiatric and neurological disorders. Here, we describe the design and synthesis of a series of (R)-3-(5-furanyl)carboxamido-2-aminopropanoic acid analogues 8a-s as agonists at the glycine (Gly) binding site in the GluN1 subunit, but not GluN3 subunits, of NMDA receptors. These novel analogues display highly variable potencies and agonist efficacies among the NMDA receptor subtypes (GluN1/2A-D) in a manner dependent on the GluN2 subunit. Notably, compound 8p is identified as a potent partial agonist at GluN1/2C (EC50 = 0.074 μM) with an agonist efficacy of 28% relative to activation by Gly and virtually no agonist activity at GluN1/2A, GluN1/2B, and GluN1/2D. Thus, these novel agonists can modulate the activity of specific NMDA receptor subtypes by replacing the full endogenous agonists Gly or d-serine (d-Ser), thereby providing new opportunities in the development of novel therapeutic agents.
Collapse
Affiliation(s)
- Fabao Zhao
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2200, Denmark.,Current address: Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong, P.R. China
| | - Unai Atxabal
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2200, Denmark
| | - Sofia Mariottini
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2200, Denmark
| | - Feng Yi
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT 59812
| | - James S. Lotti
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT 59812
| | - Nirvan Rouzbeh
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT 59812
| | - Na Liu
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2200, Denmark.,Current address: Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong, P.R. China
| | - Lennart Bunch
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2200, Denmark
| | - Kasper B. Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT 59812.,Corresponding Authors: Kasper B. Hansen - Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, United States; Phone: (+1) 4062434820; . Rasmus P. Clausen - Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark; Phone: (+45) 35336566;
| | - Rasmus P. Clausen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2200, Denmark.,Corresponding Authors: Kasper B. Hansen - Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, United States; Phone: (+1) 4062434820; . Rasmus P. Clausen - Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark; Phone: (+45) 35336566;
| |
Collapse
|
21
|
Veselinović T, Neuner I. Progress and Pitfalls in Developing Agents to Treat Neurocognitive Deficits Associated with Schizophrenia. CNS Drugs 2022; 36:819-858. [PMID: 35831706 PMCID: PMC9345797 DOI: 10.1007/s40263-022-00935-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 12/11/2022]
Abstract
Cognitive impairments associated with schizophrenia (CIAS) represent a central element of the symptomatology of this severe mental disorder. CIAS substantially determine the disease prognosis and hardly, if at all, respond to treatment with currently available antipsychotics. Remarkably, all drugs presently approved for the treatment of schizophrenia are, to varying degrees, dopamine D2/D3 receptor blockers. In turn, rapidly growing evidence suggests the immense significance of systems other than the dopaminergic system in the genesis of CIAS. Accordingly, current efforts addressing the unmet needs of patients with schizophrenia are primarily based on interventions in other non-dopaminergic systems. In this review article, we provide a brief overview of the available evidence on the importance of specific systems in the development of CIAS. In addition, we describe the promising targets for the development of new drugs that have been used so far. In doing so, we present the most important candidates that have been investigated in the field of the specific systems in recent years and present a summary of the results available at the time of drafting this review (May 2022), as well as the currently ongoing studies.
Collapse
Affiliation(s)
- Tanja Veselinović
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.
| | - Irene Neuner
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- JARA-BRAIN, Aachen, Germany
| |
Collapse
|
22
|
Hansen KB, Wollmuth LP, Bowie D, Furukawa H, Menniti FS, Sobolevsky AI, Swanson GT, Swanger SA, Greger IH, Nakagawa T, McBain CJ, Jayaraman V, Low CM, Dell'Acqua ML, Diamond JS, Camp CR, Perszyk RE, Yuan H, Traynelis SF. Structure, Function, and Pharmacology of Glutamate Receptor Ion Channels. Pharmacol Rev 2021; 73:298-487. [PMID: 34753794 PMCID: PMC8626789 DOI: 10.1124/pharmrev.120.000131] [Citation(s) in RCA: 267] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Many physiologic effects of l-glutamate, the major excitatory neurotransmitter in the mammalian central nervous system, are mediated via signaling by ionotropic glutamate receptors (iGluRs). These ligand-gated ion channels are critical to brain function and are centrally implicated in numerous psychiatric and neurologic disorders. There are different classes of iGluRs with a variety of receptor subtypes in each class that play distinct roles in neuronal functions. The diversity in iGluR subtypes, with their unique functional properties and physiologic roles, has motivated a large number of studies. Our understanding of receptor subtypes has advanced considerably since the first iGluR subunit gene was cloned in 1989, and the research focus has expanded to encompass facets of biology that have been recently discovered and to exploit experimental paradigms made possible by technological advances. Here, we review insights from more than 3 decades of iGluR studies with an emphasis on the progress that has occurred in the past decade. We cover structure, function, pharmacology, roles in neurophysiology, and therapeutic implications for all classes of receptors assembled from the subunits encoded by the 18 ionotropic glutamate receptor genes. SIGNIFICANCE STATEMENT: Glutamate receptors play important roles in virtually all aspects of brain function and are either involved in mediating some clinical features of neurological disease or represent a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of this class of receptors will advance our understanding of many aspects of brain function at molecular, cellular, and system levels and provide new opportunities to treat patients.
Collapse
Affiliation(s)
- Kasper B Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Lonnie P Wollmuth
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Derek Bowie
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Hiro Furukawa
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Frank S Menniti
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Alexander I Sobolevsky
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Geoffrey T Swanson
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Sharon A Swanger
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Ingo H Greger
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Terunaga Nakagawa
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chris J McBain
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Vasanthi Jayaraman
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chian-Ming Low
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Mark L Dell'Acqua
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Jeffrey S Diamond
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chad R Camp
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Riley E Perszyk
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Hongjie Yuan
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Stephen F Traynelis
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| |
Collapse
|
23
|
Sherwood MW, Oliet SHR, Panatier A. NMDARs, Coincidence Detectors of Astrocytic and Neuronal Activities. Int J Mol Sci 2021; 22:7258. [PMID: 34298875 PMCID: PMC8307462 DOI: 10.3390/ijms22147258] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
Synaptic plasticity is an extensively studied cellular correlate of learning and memory in which NMDARs play a starring role. One of the most interesting features of NMDARs is their ability to act as a co-incident detector. It is unique amongst neurotransmitter receptors in this respect. Co-incident detection is possible because the opening of NMDARs requires membrane depolarisation and the binding of glutamate. Opening of NMDARs also requires a co-agonist. Although the dynamic regulation of glutamate and membrane depolarization have been well studied in coincident detection, the role of the co-agonist site is unexplored. It turns out that non-neuronal glial cells, astrocytes, regulate co-agonist availability, giving them the ability to influence synaptic plasticity. The unique morphology and spatial arrangement of astrocytes at the synaptic level affords them the capacity to sample and integrate information originating from unrelated synapses, regardless of any pre-synaptic and post-synaptic commonality. As astrocytes are classically considered slow responders, their influence at the synapse is widely recognized as modulatory. The aim herein is to reconsider the potential of astrocytes to participate directly in ongoing synaptic NMDAR activity and co-incident detection.
Collapse
Affiliation(s)
- Mark W. Sherwood
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France;
| | | | - Aude Panatier
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France;
| |
Collapse
|
24
|
Inhibition of Glycine Re-Uptake: A Potential Approach for Treating Pain by Augmenting Glycine-Mediated Spinal Neurotransmission and Blunting Central Nociceptive Signaling. Biomolecules 2021; 11:biom11060864. [PMID: 34200954 PMCID: PMC8230656 DOI: 10.3390/biom11060864] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/23/2022] Open
Abstract
Among the myriad of cellular and molecular processes identified as contributing to pathological pain, disinhibition of spinal cord nociceptive signaling to higher cortical centers plays a critical role. Importantly, evidence suggests that impaired glycinergic neurotransmission develops in the dorsal horn of the spinal cord in inflammatory and neuropathic pain models and is a key maladaptive mechanism causing mechanical hyperalgesia and allodynia. Thus, it has been hypothesized that pharmacological agents capable of augmenting glycinergic tone within the dorsal horn may be able to blunt or block aberrant nociceptor signaling to the brain and serve as a novel class of analgesics for various pathological pain states. Indeed, drugs that enhance dysfunctional glycinergic transmission, and in particular inhibitors of the glycine transporters (GlyT1 and GlyT2), are generating widespread interest as a potential class of novel analgesics. The GlyTs are Na+/Cl−-dependent transporters of the solute carrier 6 (SLC6) family and it has been proposed that the inhibition of them presents a possible mechanism by which to increase spinal extracellular glycine concentrations and enhance GlyR-mediated inhibitory neurotransmission in the dorsal horn. Various inhibitors of both GlyT1 and GlyT2 have demonstrated broad analgesic efficacy in several preclinical models of acute and chronic pain, providing promise for the approach to deliver a first-in-class non-opioid analgesic with a mechanism of action differentiated from current standard of care. This review will highlight the therapeutic potential of GlyT inhibitors as a novel class of analgesics, present recent advances reported for the field, and discuss the key challenges associated with the development of a GlyT inhibitor into a safe and effective agent to treat pain.
Collapse
|
25
|
Chang CY, Luo DZ, Pei JC, Kuo MC, Hsieh YC, Lai WS. Not Just a Bystander: The Emerging Role of Astrocytes and Research Tools in Studying Cognitive Dysfunctions in Schizophrenia. Int J Mol Sci 2021; 22:ijms22105343. [PMID: 34069523 PMCID: PMC8160762 DOI: 10.3390/ijms22105343] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022] Open
Abstract
Cognitive dysfunction is one of the core symptoms in schizophrenia, and it is predictive of functional outcomes and therefore useful for treatment targets. Rather than improving cognitive deficits, currently available antipsychotics mainly focus on positive symptoms, targeting dopaminergic/serotoninergic neurons and receptors in the brain. Apart from investigating the neural mechanisms underlying schizophrenia, emerging evidence indicates the importance of glial cells in brain structure development and their involvement in cognitive functions. Although the etiopathology of astrocytes in schizophrenia remains unclear, accumulated evidence reveals that alterations in gene expression and astrocyte products have been reported in schizophrenic patients. To further investigate the role of astrocytes in schizophrenia, we highlighted recent progress in the investigation of the effect of astrocytes on abnormalities in glutamate transmission and impairments in the blood–brain barrier. Recent advances in animal models and behavioral methods were introduced to examine schizophrenia-related cognitive deficits and negative symptoms. We also highlighted several experimental tools that further elucidate the role of astrocytes. Instead of focusing on schizophrenia as a neuron-specific disorder, an additional astrocytic perspective provides novel and promising insight into its causal mechanisms and treatment. The involvement of astrocytes in the pathogenesis of schizophrenia and other brain disorders is worth further investigation.
Collapse
Affiliation(s)
- Chia-Yuan Chang
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 10617, Taiwan;
| | - Da-Zhong Luo
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
| | - Ju-Chun Pei
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
| | - Ming-Che Kuo
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 10617, Taiwan;
- Department of Neurology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Yi-Chen Hsieh
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan; (C.-Y.C.); (D.-Z.L.); (J.-C.P.); (Y.-C.H.)
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 10617, Taiwan;
- Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: ; Tel.: +886-2-3366-3112; Fax: +886-2-3362-9909
| |
Collapse
|
26
|
Mediatory role of the dopaminergic system through D1 receptor on glycine-induced hypophagia in neonatal broiler-type chickens. Amino Acids 2021; 53:461-470. [PMID: 33649971 DOI: 10.1007/s00726-021-02963-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 02/19/2021] [Indexed: 10/22/2022]
Abstract
The present study aimed to examine the mediatory role of the dopaminergic system in the food intake induced by intracerebroventricular (ICV) injection of glycine in neonatal 3-h feed-deprived (FD3) meat-type chickens. In the first and second experiments, birds were ICV injected using low and high doses of glycine (50, 100 and 200 nmol) and strychnine (50, 100 and 200 nmol), respectively. In experiments 3-9, the behaviorally subeffective doses of dopamine (10 nmol), 6-OHDA (2.5 nmol), SCH 23,390 (D1 antagonist; 5 nmol), AMI-193 (D2 antagonist; 5 nmol), NGB2904 (D3 antagonist; 6.4 nmol) and L-741,742 (D4 antagonist; 6 nmol) were, respectively, co-administrated with glycine (200 nmol) in FD3 5-day-old chicks to investigate possible interplay of dopamine receptors in glycine-induced feeding behavior. Then, cumulative food intake based on body weight percentage (%BW) was determined at 30, 60 and 120 min after the injection. According to the results, dopamine significantly boosted the hypophagia induced by glycine at all-time intervals (p ≤ 0.001). These results combined with the previous findings suggest an interplay between dopamine and glycine in chicken's brain in which D1 receptor-mediated food intake induced by glycine.
Collapse
|
27
|
Pei JC, Luo DZ, Gau SS, Chang CY, Lai WS. Directly and Indirectly Targeting the Glycine Modulatory Site to Modulate NMDA Receptor Function to Address Unmet Medical Needs of Patients With Schizophrenia. Front Psychiatry 2021; 12:742058. [PMID: 34658976 PMCID: PMC8517243 DOI: 10.3389/fpsyt.2021.742058] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 12/30/2022] Open
Abstract
Schizophrenia is a severe mental illness that affects ~1% of the world's population. It is clinically characterized by positive, negative, and cognitive symptoms. Currently available antipsychotic medications are relatively ineffective in improving negative and cognitive deficits, which are related to a patient's functional outcomes and quality of life. Negative symptoms and cognitive deficits are unmet by the antipsychotic medications developed to date. In recent decades, compelling animal and clinical studies have supported the NMDA receptor (NMDAR) hypofunction hypothesis of schizophrenia and have suggested some promising therapeutic agents. Notably, several NMDAR-enhancing agents, especially those that function through the glycine modulatory site (GMS) of NMDAR, cause significant reduction in psychotic and cognitive symptoms in patients with schizophrenia. Given that the NMDAR-mediated signaling pathway has been implicated in cognitive/social functions and that GMS is a potential therapeutic target for enhancing the activation of NMDARs, there is great interest in investigating the effects of direct and indirect GMS modulators and their therapeutic potential. In this review, we focus on describing preclinical and clinical studies of direct and indirect GMS modulators in the treatment of schizophrenia, including glycine, D-cycloserine, D-serine, glycine transporter 1 (GlyT1) inhibitors, and D-amino acid oxidase (DAO or DAAO) inhibitors. We highlight some of the most promising recently developed pharmacological compounds designed to either directly or indirectly target GMS and thus augment NMDAR function to treat the cognitive and negative symptoms of schizophrenia. Overall, the current findings suggest that indirectly targeting of GMS appears to be more beneficial and leads to less adverse effects than direct targeting of GMS to modulate NMDAR functions. Indirect GMS modulators, especially GlyT1 inhibitors and DAO inhibitors, open new avenues for the treatment of unmet medical needs for patients with schizophrenia.
Collapse
Affiliation(s)
- Ju-Chun Pei
- Department of Psychology, National Taiwan University, Taipei, Taiwan
| | - Da-Zhong Luo
- Department of Psychology, National Taiwan University, Taipei, Taiwan
| | - Shiang-Shin Gau
- Department of Psychology, National Taiwan University, Taipei, Taiwan
| | - Chia-Yuan Chang
- Department of Psychology, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
28
|
Nagy LV, Bali ZK, Kapus G, Pelsőczi P, Farkas B, Lendvai B, Lévay G, Hernádi I. Converging Evidence on D-Amino Acid Oxidase-Dependent Enhancement of Hippocampal Firing Activity and Passive Avoidance Learning in Rats. Int J Neuropsychopharmacol 2020; 24:434-445. [PMID: 33305805 PMCID: PMC8130201 DOI: 10.1093/ijnp/pyaa095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND N-methyl-D-aspartate (NMDA) receptor activation requires the binding of a co-agonist on the glycine-binding site. D-serine is the main endogenous co-agonist of NMDA receptors, and its availability significantly depends on the activity of the metabolic enzyme D-amino acid oxidase (DAAO). Inhibition of DAAO increases the brain levels of D-serine and modulates a variety of physiological functions, including cognitive behavior. METHODS Here, we examined the effects of a novel 4-hydroxypyridazin-3(2H)-one derivative DAAO inhibitor, Compound 30 (CPD30), on passive avoidance learning and on neuronal firing activity in rats. RESULTS D-serine administration was applied as reference, which increased cognitive performance and enhanced hippocampal firing activity and responsiveness to NMDA after both local and systemic application. Similarly to D-serine, CPD30 (0.1 mg/kg) effectively reversed MK-801-induced memory impairment in the passive avoidance test. Furthermore, local iontophoretic application of CPD30 in the vicinity of hippocampal pyramidal neurons significantly increased firing rate and enhanced their responses to locally applied NMDA. CPD30 also enhanced hippocampal firing activity after systemic administration. In 0.1- to 1.0-mg/kg doses, CPD30 increased spontaneous and NMDA-evoked firing activity of the neurons. Effects of CPD30 on NMDA responsiveness emerged faster (at 10 minutes post-injection) when a 1.0-mg/kg dose was applied compared with the onset of the effects of 0.1 mg/kg CPD30 (at 30 minutes post-injection). CONCLUSIONS The present results confirm that the inhibition of DAAO enzyme is an effective strategy for cognitive enhancement. Our findings further facilitate the understanding of the cellular mechanisms underlying the behavioral effects of DAAO inhibition in the mammalian brain.
Collapse
Affiliation(s)
- Lili Veronika Nagy
- Department of Experimental Zoology and Neurobiology, Faculty of Sciences, University of Pécs, Pécs, Hungary,Szentágothai Research Center, Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Zsolt Kristóf Bali
- Szentágothai Research Center, Center for Neuroscience, University of Pécs, Pécs, Hungary,Grastyán Translational Research Center, University of Pécs, Pécs, Hungary,Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary,Correspondence: Zsolt Kristóf Bali, PhD, Translational Neuroscience Research Group, Grastyán Translational Research Center, 6 Ifjúság út, H-7624, Pécs, Hungary ()
| | - Gábor Kapus
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Péter Pelsőczi
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Bence Farkas
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Balázs Lendvai
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - György Lévay
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary,Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - István Hernádi
- Department of Experimental Zoology and Neurobiology, Faculty of Sciences, University of Pécs, Pécs, Hungary,Szentágothai Research Center, Center for Neuroscience, University of Pécs, Pécs, Hungary,Grastyán Translational Research Center, University of Pécs, Pécs, Hungary,Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
29
|
Coyle JT, Ruzicka WB, Balu DT. Fifty Years of Research on Schizophrenia: The Ascendance of the Glutamatergic Synapse. Am J Psychiatry 2020; 177:1119-1128. [PMID: 33256439 PMCID: PMC8011846 DOI: 10.1176/appi.ajp.2020.20101481] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
30
|
Candidate metabolic biomarkers for schizophrenia in CNS and periphery: Do any possible associations exist? Schizophr Res 2020; 226:95-110. [PMID: 30935700 DOI: 10.1016/j.schres.2019.03.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 02/07/2023]
Abstract
Due to the limitations of analytical techniques and the complicity of schizophrenia, nowadays it is still a challenge to diagnose and stratify schizophrenia patients accurately. Many attempts have been made to identify and validate available biomarkers for schizophrenia from CSF and/or peripheral blood in clinical studies with consideration to disease stages, antipsychotic effects and even gender differences. However, conflicting results handicap the validation and application of biomarkers for schizophrenia. In view of availability and feasibility, peripheral biomarkers have superior advantages over biomarkers in CNS. Meanwhile, schizophrenia is considered to be a devastating neuropsychiatric disease mainly taking place in CNS featured by widespread defects in multiple metabolic pathways whose dynamic interactions, until recently, have been difficult to difficult to investigate. Evidence for these alterations has been collected piecemeal, limiting the potential to inform our understanding of the interactions among relevant biochemical pathways. Taken these points together, it will be interesting to investigate possible associations of biomarkers between CNS and periphery. Numerous studies have suggested putative correlations within peripheral and CNS systems especially for dopaminergic and glutamatergic metabolic biomarkers. In addition, it has been demonstrated that blood concentrations of BDNF protein can also reflect its changes in the nervous system. In turn, BDNF also interacts with glutamatergic, dopaminergic and serotonergic systems. Therefore, this review will summarize metabolic biomarkers identified both in the CNS (brain tissues and CSF) and peripheral blood. Further, more attentions will be paid to discussing possible physical and functional associations between CNS and periphery, especially with respect to BDNF.
Collapse
|
31
|
Sheipouri D, Gallagher CI, Shimmon S, Rawling T, Vandenberg RJ. A System for Assessing Dual Action Modulators of Glycine Transporters and Glycine Receptors. Biomolecules 2020; 10:E1618. [PMID: 33266066 PMCID: PMC7760315 DOI: 10.3390/biom10121618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
Reduced inhibitory glycinergic neurotransmission is implicated in a number of neurological conditions such as neuropathic pain, schizophrenia, epilepsy and hyperekplexia. Restoring glycinergic signalling may be an effective method of treating these pathologies. Glycine transporters (GlyTs) control synaptic and extra-synaptic glycine concentrations and slowing the reuptake of glycine using specific GlyT inhibitors will increase glycine extracellular concentrations and increase glycine receptor (GlyR) activation. Glycinergic neurotransmission can also be improved through positive allosteric modulation (PAM) of GlyRs. Despite efforts to manipulate this synapse, no therapeutics currently target it. We propose that dual action modulators of both GlyTs and GlyRs may show greater therapeutic potential than those targeting individual proteins. To show this, we have characterized a co-expression system in Xenopus laevis oocytes consisting of GlyT1 or GlyT2 co-expressed with GlyRα1. We use two electrode voltage clamp recording techniques to measure the impact of GlyTs on GlyRs and the effects of modulators of these proteins. We show that increases in GlyT density in close proximity to GlyRs diminish receptor currents. Reductions in GlyR mediated currents are not observed when non-transportable GlyR agonists are applied or when Na+ is not available. GlyTs reduce glycine concentrations across different concentration ranges, corresponding with their ion-coupling stoichiometry, and full receptor currents can be restored when GlyTs are blocked with selective inhibitors. We show that partial inhibition of GlyT2 and modest GlyRα1 potentiation using a dual action compound, is as useful in restoring GlyR currents as a full and potent single target GlyT2 inhibitor or single target GlyRα1 PAM. The co-expression system developed in this study will provide a robust means for assessing the likely impact of GlyR PAMs and GlyT inhibitors on glycine neurotransmission.
Collapse
Affiliation(s)
- Diba Sheipouri
- School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia; (D.S.); (C.I.G.)
| | - Casey I. Gallagher
- School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia; (D.S.); (C.I.G.)
| | - Susan Shimmon
- School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (S.S.); (T.R.)
| | - Tristan Rawling
- School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (S.S.); (T.R.)
| | - Robert J. Vandenberg
- School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia; (D.S.); (C.I.G.)
| |
Collapse
|
32
|
Postsynaptic Serine Racemase Regulates NMDA Receptor Function. J Neurosci 2020; 40:9564-9575. [PMID: 33158959 DOI: 10.1523/jneurosci.1525-20.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/23/2020] [Accepted: 11/01/2020] [Indexed: 01/03/2023] Open
Abstract
d-serine is the primary NMDAR coagonist at mature forebrain synapses and is synthesized by the enzyme serine racemase (SR). However, our understanding of the mechanisms regulating the availability of synaptic d-serine remains limited. Though early studies suggested d-serine is synthesized and released from astrocytes, more recent studies have demonstrated a predominantly neuronal localization of SR. More specifically, recent work intriguingly suggests that SR may be found at the postsynaptic density, yet the functional implications of postsynaptic SR on synaptic transmission are not yet known. Here, we show an age-dependent dendritic and postsynaptic localization of SR and d-serine by immunohistochemistry and electron microscopy in mouse CA1 pyramidal neurons. In addition, using a single-neuron genetic approach in SR conditional KO mice from both sexes, we demonstrate a cell-autonomous role for SR in regulating synaptic NMDAR function at Schaffer collateral (CA3)-CA1 synapses. Importantly, single-neuron genetic deletion of SR resulted in the elimination of LTP at 1 month of age, which could be rescued by exogenous d-serine. Interestingly, there was a restoration of LTP by 2 months of age that was associated with an upregulation of synaptic GluN2B. Our findings support a cell-autonomous role for postsynaptic neuronal SR in regulating synaptic NMDAR function and suggests a possible autocrine mode of d-serine action.SIGNIFICANCE STATEMENT NMDARs are key regulators of neurodevelopment and synaptic plasticity and are unique in their requirement for binding of a coagonist, which is d-serine at most forebrain synapses. However, our understanding of the mechanisms regulating synaptic d-serine availability remains limited. d-serine is synthesized in the brain by the neuronal enzyme serine racemase (SR). Here, we show dendritic and postsynaptic localization of SR and d-serine in CA1 pyramidal neurons. In addition, using single-neuron genetic deletion of SR, we establish a role of postsynaptic SR in regulating NMDAR function. These results support an autocrine mode of d-serine action at synapses.
Collapse
|
33
|
Kantak KM, Gauthier JM, Mathieson E, Knyazhanskaya E, Rodriguez-Echemendia P, Man HY. Sex differences in the effects of a combined behavioral and pharmacological treatment strategy for cocaine relapse prevention in an animal model of cue exposure therapy. Behav Brain Res 2020; 395:112839. [PMID: 32750464 DOI: 10.1016/j.bbr.2020.112839] [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: 04/27/2020] [Revised: 07/05/2020] [Accepted: 07/26/2020] [Indexed: 10/23/2022]
Abstract
Brief interventions of environmental enrichment (EE) or the glycine transporter-1 inhibitor Org24598 administered with cocaine-cue extinction training were shown previously to inhibit reacquisition of cocaine self-administration in male rats trained to self-administer a moderate 0.3 mg/kg dose of cocaine. Determining how EE and Org24598 synergize in combination in an animal model of cue exposure therapy is novel. Important changes made in this investigation were increasing the cocaine training dose to 1.0 mg/kg and determining sex differences. Adult male and female rats self-administering 1.0 mg/kg cocaine for 35-40 daily sessions exhibited an addiction-like phenotype under a second-order schedule of cocaine delivery and cue presentation. Rats next underwent 6 weekly extinction training sessions for which treatments consisted of EE or NoEE and Vehicle or Org24598 (3.0 mg/kg in males; 3.0 or 7.5 mg/kg in females). Rats then were tested for reacquisition of cocaine self-administration for 15 daily sessions. In males, the combined EE +3.0 mg/kg Org24598 treatment facilitated extinction learning and inhibited reacquisition of cocaine self-administration to a greater extent than no treatment and to individual EE or 3.0 mg/kg Org24598 treatments. In females, EE +7.5 mg/kg Org24598 facilitated extinction learning, but did not inhibit reacquisition of cocaine self-administration. Thus, there were sex differences in the ability of EE + Org24598 administered in conjunction with extinction training to inhibit cocaine relapse in rats exhibiting an addiction-like phenotype. These findings suggest that this multimodal treatment approach might be a feasible option during cue exposure therapy in cocaine-dependent men, but not women.
Collapse
Affiliation(s)
- Kathleen M Kantak
- Department of Psychological and Brain Sciences, Boston University, Boston, USA; Center for Systems Neuroscience, Boston University, Boston, USA.
| | - Jamie M Gauthier
- Department of Psychological and Brain Sciences, Boston University, Boston, USA
| | - Elon Mathieson
- Department of Psychological and Brain Sciences, Boston University, Boston, USA
| | | | | | - Heng-Ye Man
- Department of Biology, Boston University, Boston, USA; Center for Systems Neuroscience, Boston University, Boston, USA
| |
Collapse
|
34
|
Wolosker H, Balu DT. D-Serine as the gatekeeper of NMDA receptor activity: implications for the pharmacologic management of anxiety disorders. Transl Psychiatry 2020; 10:184. [PMID: 32518273 PMCID: PMC7283225 DOI: 10.1038/s41398-020-00870-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/04/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
Fear, anxiety, and trauma-related disorders, including post-traumatic stress disorder (PTSD), are quite common and debilitating, with an estimated lifetime prevalence of ~28% in Western populations. They are associated with excessive fear reactions, often including an inability to extinguish learned fear, increased avoidance behavior, as well as altered cognition and mood. There is an extensive literature demonstrating the importance of N-methyl-D-aspartate receptor (NMDAR) function in regulating these behaviors. NMDARs require the binding of a co-agonist, D-serine or glycine, at the glycine modulatory site (GMS) to function. D-serine is now garnering attention as the primary NMDAR co-agonist in limbic brain regions implicated in neuropsychiatric disorders. L-serine is synthesized by astrocytes, which is then transported to neurons for conversion to D-serine by serine racemase (SR), a model we term the 'serine shuttle.' The neuronally-released D-serine is what regulates NMDAR activity. Our review discusses how the systems that regulate the synaptic availability of D-serine, a critical gatekeeper of NMDAR-dependent activation, could be targeted to improve the pharmacologic management of anxiety-related disorders where the desired outcomes are the facilitation of fear extinction, as well as mood and cognitive enhancement.
Collapse
Affiliation(s)
- Herman Wolosker
- grid.6451.60000000121102151Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096 Israel
| | - Darrick T. Balu
- grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA 02115 USA ,grid.240206.20000 0000 8795 072XTranslational Psychiatry Laboratory, McLean Hospital, Belmont, MA 02478 USA
| |
Collapse
|
35
|
Jafari Z, Kolb BE, Mohajerani MH. Prepulse inhibition of the acoustic startle reflex and P50 gating in aging and alzheimer's disease. Ageing Res Rev 2020; 59:101028. [PMID: 32092463 DOI: 10.1016/j.arr.2020.101028] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 01/20/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023]
Abstract
Inhibition plays a crucial role in many functional domains, such as cognition, emotion, and actions. Studies on cognitive aging demonstrate changes in inhibitory mechanisms are age- and pathology-related. Prepulse inhibition (PPI) is the suppression of an acoustic startle reflex (ASR) to an intense stimulus when a weak prepulse stimulus precedes the startle stimulus. A reduction of PPI is thought to reflect dysfunction of sensorimotor gating which normally suppresses excessive behavioral responses to disruptive stimuli. Both human and rodent studies show age-dependent alterations of PPI of the ASR that are further compromised in Alzheimer's disease (AD). The auditory P50 gating, an index of repetition suppression, also is characterized as a putative electrophysiological biomarker of prodromal AD. This review provides the latest evidence of age- and AD-associated impairment of sensorimotor gating based upon both human and rodent studies, as well as the AD-related disruption of P50 gating in humans. It begins with a concise review of neural networks underlying PPI regulation. Then, evidence of age- and AD-related dysfunction of both PPI and P50 gating is discussed. The attentional/ emotional aspects of sensorimotor gating and the neurotransmitter mechanisms underpinning PPI and P50 gating are also reviewed. The review ends with conclusions and research directions.
Collapse
Affiliation(s)
- Zahra Jafari
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, T1K 3M4 AB, Canada; Department of Basic Sciences in Rehabilitation, School of Rehabilitation Sciences, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Bryan E Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, T1K 3M4 AB, Canada.
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, T1K 3M4 AB, Canada.
| |
Collapse
|
36
|
Abstract
The objective in managing patients with schizophrenia is to provide effective control of symptoms and enable the patient to reintegrate into society. Pharmacotherapy should, therefore, aim to provide optimum symptom control with minimal side effects using a simple dosage regimen. Ideally, this would be achieved through the use of a single agent. Some patients are successfully managed with a single antipsychotic drug, but it is often necessary or thought to be necessary to use a combination of agents to provide effective treatment. One European survey reported that most patients receive two to three psychotropic agents on average, but at least 5-22% receive five or more agents [53]. Unfortunately, the addition of more agents increases the risk of drug interactions, adverse events and non-compliance. This is amplified by the paucity of evidence-based medicine currently available to guide physicians in the use of combinations of agents, and the tendency of polypharmaceutical practice to be mostly driven by personal preference, clinical experience and marketing. This article therefore briefly looks at the feasibility of using various drug classes as adjunctive therapy in patients with schizophrenia.
Collapse
|
37
|
D-Serine, the Shape-Shifting NMDA Receptor Co-agonist. Neurochem Res 2020; 45:1344-1353. [PMID: 32189130 DOI: 10.1007/s11064-020-03014-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
Abstract
Shape-shifting, a phenomenon wide-spread in folklore, refers to the ability to physically change from one identity to another, typically from an innocuous entity to a destructive one. The amino acid D-serine over the last 25 years has "shape-shifted" into several identities: a purported glial transmitter activating N-methyl-D-aspartate receptors (NMDARs), a co-transmitter concentrated in excitatory glutamatergic neurons, an autocrine that is released at dendritic spines to prime their post-synaptic NMDARs for an instantaneous response to glutamate and an excitotoxic moiety released from inflammatory (A1) astrocytes. This article will review evidence in support of these scenarios and the artifacts that misled investigators of the true identity of D-serine.
Collapse
|
38
|
de Bartolomeis A, Manchia M, Marmo F, Vellucci L, Iasevoli F, Barone A. Glycine Signaling in the Framework of Dopamine-Glutamate Interaction and Postsynaptic Density. Implications for Treatment-Resistant Schizophrenia. Front Psychiatry 2020; 11:369. [PMID: 32477178 PMCID: PMC7240307 DOI: 10.3389/fpsyt.2020.00369] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Treatment-resistant schizophrenia (TRS) or suboptimal response to antipsychotics affects almost 30% of schizophrenia (SCZ) patients, and it is a relevant clinical issue with significant impact on the functional outcome and on the global burden of disease. Among putative novel treatments, glycine-centered therapeutics (i.e. sarcosine, glycine itself, D-Serine, and bitopertin) have been proposed, based on a strong preclinical rationale with, however, mixed clinical results. Therefore, a better appraisal of glycine interaction with the other major players of SCZ pathophysiology and specifically in the framework of dopamine - glutamate interactions is warranted. New methodological approaches at cutting edge of technology and drug discovery have been applied to study the role of glycine in glutamate signaling, both at presynaptic and post-synaptic level and have been instrumental for unveiling the role of glycine in dopamine-glutamate interaction. Glycine is a non-essential amino acid that plays a critical role in both inhibitory and excitatory neurotransmission. In caudal areas of central nervous system (CNS), such as spinal cord and brainstem, glycine acts as a powerful inhibitory neurotransmitter through binding to its receptor, i.e. the Glycine Receptor (GlyR). However, glycine also works as a co-agonist of the N-Methyl-D-Aspartate receptor (NMDAR) in excitatory glutamatergic neurotransmission. Glycine concentration in the synaptic cleft is finely tuned by glycine transporters, i.e. GlyT1 and GlyT2, that regulate the neurotransmitter's reuptake, with the first considered a highly potential target for psychosis therapy. Reciprocal regulation of dopamine and glycine in forebrain, glycine modulation of glutamate, glycine signaling interaction with postsynaptic density proteins at glutamatergic synapse, and human genetics of glycinergic pathways in SCZ are tackled in order to highlight the exploitation of this neurotransmitters and related molecules in SCZ and TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular Psychiatry and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Napoli Federico II, Naples, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy.,Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Federica Marmo
- Laboratory of Molecular Psychiatry and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Napoli Federico II, Naples, Italy
| | - Licia Vellucci
- Laboratory of Molecular Psychiatry and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Napoli Federico II, Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular Psychiatry and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Napoli Federico II, Naples, Italy
| | - Annarita Barone
- Laboratory of Molecular Psychiatry and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Napoli Federico II, Naples, Italy
| |
Collapse
|
39
|
Metabolomics methods to analyze full spectrum of amino acids in different domains of bovine colostrum and mature milk. Eur Food Res Technol 2019. [DOI: 10.1007/s00217-019-03385-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
40
|
Baliova M, Jursky F. Phosphomimetic Mutation of Glycine Transporter GlyT1 C-Terminal PDZ Binding Motif Inhibits its Interactions with PSD95. J Mol Neurosci 2019; 70:488-493. [DOI: 10.1007/s12031-019-01435-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/04/2019] [Indexed: 12/11/2022]
|
41
|
Wang S, Dong G, Sheng C. Structural simplification: an efficient strategy in lead optimization. Acta Pharm Sin B 2019; 9:880-901. [PMID: 31649841 PMCID: PMC6804494 DOI: 10.1016/j.apsb.2019.05.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/04/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
The trend toward designing large hydrophobic molecules for lead optimization is often associated with poor drug-likeness and high attrition rates in drug discovery and development. Structural simplification is a powerful strategy for improving the efficiency and success rate of drug design by avoiding "molecular obesity". The structural simplification of large or complex lead compounds by truncating unnecessary groups can not only improve their synthetic accessibility but also improve their pharmacokinetic profiles, reduce side effects and so on. This review will summarize the application of structural simplification in lead optimization. Numerous case studies, particularly those involving successful examples leading to marketed drugs or drug-like candidates, will be introduced and analyzed to illustrate the design strategies and guidelines for structural simplification.
Collapse
Key Words
- 11β-HSD, 11β-hydroxysteroid dehydrogenase
- 3D, three-dimensional
- ADMET, absorption, distribution, metabolism, excretion and toxicity
- AM2, adrenomedullin-2 receptor
- BIOS, biology-oriented synthesis
- CCK, cholecystokinin receptor
- CGRP, calcitonin gene-related peptide
- Drug design
- Drug discovery
- GlyT1, glycine transport 1
- HBV, hepatitis B virus
- HDAC, histone deacetylase
- HLM, human liver microsome
- JAKs, Janus tyrosine kinases
- LE, ligand efficiency
- Lead optimization
- LeuRS, leucyl-tRNA synthetase
- MCRs, multicomponent reactions
- MDR-TB, multidrug-resistant tuberculosis
- MW, molecular weight
- NP, natural product
- NPM, nucleophosmin
- PD, pharmacodynamic
- PK, pharmacokinetic
- PKC, protein kinase C
- Pharmacophore-based simplification
- Reducing chiral centers
- Reducing rings number
- SAHA, vorinostat
- SAR, structure‒activity relationship
- SCONP, structural classification of natural product
- Structural simplification
- Structure-based simplification
- TSA, trichostatin A
- TbLeuRS, T. brucei LeuRS
- ThrRS, threonyl-tRNA synthetase
- VANGL1, van-Gogh-like receptor protein 1
- aa-AMP, aminoacyl-AMP
- aa-AMS, aminoacylsulfa-moyladenosine
- aaRSs, aminoacyl-tRNA synthetases
- hA3 AR, human A3 adenosine receptor
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Shengzheng Wang
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Guoqiang Dong
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Chunquan Sheng
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
42
|
Destreel G, Seutin V, Engel D. Subsaturation of the N-methyl-D-aspartate receptor glycine site allows the regulation of bursting activity in juvenile rat nigral dopamine neurons. Eur J Neurosci 2019; 50:3454-3471. [PMID: 31206829 DOI: 10.1111/ejn.14491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/23/2019] [Accepted: 06/10/2019] [Indexed: 01/09/2023]
Abstract
The activation of N-methyl-D-aspartate receptors (NMDARs) in substantia nigra pars compacta (SNc) dopamine (DA) cells is central to generate the bursting activity, a phasic signal linked to DA-related behaviours via the change in postsynaptic DA release. NMDARs are recruited during excitatory synaptic transmission by glutamate release, but the glycine site level of occupancy of these receptors during basal action potential-dependent activity is not known for SNc DA neurons. We explored NMDAR-dependent signals during exogenous applications of co-agonists in midbrain slices from juvenile rats. We found that both glycine and D-serine strengthened the NMDAR-dependent component of excitatory postsynaptic currents (EPSCs) in a concentration-dependent manner. EPSCs were also increased by endogenous glycine via the blockade of the glycine transport. The glycine site of NMDARs contributing to synaptic transmission is therefore subsaturated. The behaviourally relevant burst firing was more sensitive to exogenous D-serine and endogenous glycine than to exogenous glycine. The mechanisms regulating the availability of the co-agonists exert consequently a critical influence on the excitability of DA neurons via NMDARs. The modulation of the phasic firing in DA neurons by ambient NMDAR co-agonists may be important for nigral information processing and downstream motor-related behaviour.
Collapse
Affiliation(s)
- Geoffrey Destreel
- GIGA-Neurosciences, Neurophysiology Group, University of Liege, Liege, Belgium
| | - Vincent Seutin
- GIGA-Neurosciences, Neurophysiology Group, University of Liege, Liege, Belgium
| | - Dominique Engel
- GIGA-Neurosciences, Neurophysiology Group, University of Liege, Liege, Belgium
| |
Collapse
|
43
|
Erdem FA, Ilic M, Koppensteiner P, Gołacki J, Lubec G, Freissmuth M, Sandtner W. A comparison of the transport kinetics of glycine transporter 1 and glycine transporter 2. J Gen Physiol 2019; 151:1035-1050. [PMID: 31270129 PMCID: PMC6683666 DOI: 10.1085/jgp.201912318] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/16/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022] Open
Abstract
Erdem et al. compare the kinetics of the SLC6 family glycine transporters GlyT1 and GlyT2. Though the two transporters are rate-limited by distinct reaction steps, they both display high transport capacity, with the kinetics of GlyT1 sufficient to supply extracellular glycine to the NMDA receptor. Transporters of the solute carrier 6 (SLC6) family translocate their cognate substrate together with Na+ and Cl−. Detailed kinetic models exist for the transporters of GABA (GAT1/SLC6A1) and the monoamines dopamine (DAT/SLC6A3) and serotonin (SERT/SLC6A4). Here, we posited that the transport cycle of individual SLC6 transporters reflects the physiological requirements they operate under. We tested this hypothesis by analyzing the transport cycle of glycine transporter 1 (GlyT1/SLC6A9) and glycine transporter 2 (GlyT2/SLC6A5). GlyT2 is the only SLC6 family member known to translocate glycine, Na+, and Cl− in a 1:3:1 stoichiometry. We analyzed partial reactions in real time by electrophysiological recordings. Contrary to monoamine transporters, both GlyTs were found to have a high transport capacity driven by rapid return of the empty transporter after release of Cl− on the intracellular side. Rapid cycling of both GlyTs was further supported by highly cooperative binding of cosubstrate ions and substrate such that their forward transport mode was maintained even under conditions of elevated intracellular Na+ or Cl−. The most important differences in the transport cycle of GlyT1 and GlyT2 arose from the kinetics of charge movement and the resulting voltage-dependent rate-limiting reactions: the kinetics of GlyT1 were governed by transition of the substrate-bound transporter from outward- to inward-facing conformations, whereas the kinetics of GlyT2 were governed by Na+ binding (or a related conformational change). Kinetic modeling showed that the kinetics of GlyT1 are ideally suited for supplying the extracellular glycine levels required for NMDA receptor activation.
Collapse
Affiliation(s)
- Fatma Asli Erdem
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marija Ilic
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | | | - Jakub Gołacki
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gert Lubec
- Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Glycine Metabolism and Its Alterations in Obesity and Metabolic Diseases. Nutrients 2019; 11:nu11061356. [PMID: 31208147 PMCID: PMC6627940 DOI: 10.3390/nu11061356] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/07/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
Glycine is the proteinogenic amino-acid of lowest molecular weight, harboring a hydrogen atom as a side-chain. In addition to being a building-block for proteins, glycine is also required for multiple metabolic pathways, such as glutathione synthesis and regulation of one-carbon metabolism. Although generally viewed as a non-essential amino-acid, because it can be endogenously synthesized to a certain extent, glycine has also been suggested as a conditionally essential amino acid. In metabolic disorders associated with obesity, type 2 diabetes (T2DM), and non-alcoholic fatty liver disease (NAFLDs), lower circulating glycine levels have been consistently observed, and clinical studies suggest the existence of beneficial effects induced by glycine supplementation. The present review aims at synthesizing the recent advances in glycine metabolism, pinpointing its main metabolic pathways, identifying the causes leading to glycine deficiency-especially in obesity and associated metabolic disorders-and evaluating the potential benefits of increasing glycine availability to curb the progression of obesity and obesity-related metabolic disturbances. This study focuses on the importance of diet, gut microbiota, and liver metabolism in determining glycine availability in obesity and associated metabolic disorders.
Collapse
|
45
|
Uno Y, Coyle JT. Glutamate hypothesis in schizophrenia. Psychiatry Clin Neurosci 2019; 73:204-215. [PMID: 30666759 DOI: 10.1111/pcn.12823] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/13/2022]
Abstract
Schizophrenia is a chronic and severe psychiatric disorder that has profound impact on an individual's life and on society. Thus, developing more effective therapeutic interventions is essential. Over the past quarter-century, an abundance of evidence from pharmacologic challenges, post-mortem studies, brain imaging, and genetic studies supports the role of glutamatergic dysregulation in the pathophysiology of schizophrenia, and the results of recent randomized clinical trials based on this evidence have yielded promising results. In this article, we review the evidence that alterations in glutamatergic neurotransmission, especially focusing on the N-methyl-d-aspartate receptor (NMDAR) function, may be a critical causative feature of schizophrenia, how this contributes to pathologic circuit function in the brain, and how these insights are revealing whole new avenues for treatment development that could reduce treatment-resistant symptoms, which account for persistent disability.
Collapse
Affiliation(s)
- Yota Uno
- Department of Psychiatry, Harvard Medical School, Boston, USA.,Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, USA.,Department of Psychology, University of Bath, Bath, UK
| | - Joseph T Coyle
- Department of Psychiatry, Harvard Medical School, Boston, USA.,Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, USA
| |
Collapse
|
46
|
Dantsuji M, Nakamura S, Nakayama K, Mochizuki A, Park SK, Bae YC, Ozeki M, Inoue T. 5-HT 2A receptor activation enhances NMDA receptor-mediated glutamate responses through Src kinase in the dendrites of rat jaw-closing motoneurons. J Physiol 2019; 597:2565-2589. [PMID: 30919966 DOI: 10.1113/jp275440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/26/2019] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS 5-HT increases the excitability of brainstem and spinal motoneurons, including the jaw-closing motoneurons, by depolarizing the membrane potential and decreasing the medium-duration afterhyperpolarization. In this study, we focused on how 5-HT enhances postsynaptic glutamatergic responses in the dendrites of the jaw-closing motoneurons. We demonstrate that 5-HT augments glutamatergic signalling by enhancing the function of the GluN2A-containing NMDA receptor (NMDAR) through the activation of 5-HT2A receptors (5-HT2A Rs) and Src kinase. To enhance glutamatergic responses, activation of the 5-HT2A Rs must occur within ∼60 μm of the location of the glutamate responses. 5-HT inputs to the jaw-closing motoneurons can significantly vary their input-output relationship, which may contribute to wide-range regulation of contractile forces of the jaw-closing muscles. ABSTRACT Various motor behaviours are modulated by 5-HT. Although the masseter (jaw-closing) motoneurons receive both glutamatergic and serotonergic inputs, it remains unclear how 5-HT affects the glutamatergic inputs to the motoneuronal dendrites. We examined the effects of 5-HT on postsynaptic responses evoked by single- or two-photon uncaging of caged glutamate (glutamate responses) to the dendrites of masseter motoneurons in postnatal day 2-5 rats of either sex. Application of 5-HT induced membrane depolarization and enhanced the glutamate-response amplitude. This enhancement was mimicked by the 5-HT2A receptor (5-HT2A R) agonist and was blocked by the 5-HT2A/2C R antagonist. However, neither the 5-HT2B R nor the 5-HT2C R agonists altered glutamate responses. Blockade of the NMDA receptors (NMDARs), but not AMPA receptors, abolished the 5-HT-induced enhancement. Furthermore, the selective antagonist for the GluN2A subunit abolished the 5-HT-induced enhancement. 5-HT increased GluN2A phosphorylation, while the Src kinase inhibitor reduced the 5-HT-induced enhancement and GluN2A phosphorylation. When exposure to the 5-HT2A R agonist was targeted to the dendrites, the enhancement of glutamate responses was restricted to the loci of the dendrites near the puff loci. Electron microscopic immunohistochemistry revealed that both the NMDARs and the 5-HT2A Rs were close to each other in the same dendrite. These results suggest that activation of dendritic 5-HT2A Rs enhances the function of local GluN2A-containing NMDARs through Src kinase. Such enhancement of the glutamate responses by 5-HT may contribute to wide-range regulation of contractile forces of the jaw-closing muscles.
Collapse
Affiliation(s)
- Masanori Dantsuji
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan.,Department of Implant Dentistry, Showa University School of Dentistry, Oota-ku, Tokyo, 145-8515, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Kiyomi Nakayama
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Ayako Mochizuki
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Sook Kyung Park
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 700-412, Republic of Korea
| | - Yong Chul Bae
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 700-412, Republic of Korea
| | - Masahiko Ozeki
- Department of Implant Dentistry, Showa University School of Dentistry, Oota-ku, Tokyo, 145-8515, Japan
| | - Tomio Inoue
- Department of Oral Physiology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, 142-8555, Japan
| |
Collapse
|
47
|
Zafra F, Ibáñez I, Bartolomé-Martín D, Piniella D, Arribas-Blázquez M, Giménez C. Glycine Transporters and Its Coupling with NMDA Receptors. ADVANCES IN NEUROBIOLOGY 2018; 16:55-83. [PMID: 28828606 DOI: 10.1007/978-3-319-55769-4_4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glycine plays two roles in neurotransmission. In caudal areas like the spinal cord and the brainstem, it acts as an inhibitory neurotransmitter, but in all regions of the CNS, it also works as a co-agonist with L-glutamate at N-methyl-D-aspartate receptors (NMDARs). The glycine fluxes in the CNS are regulated by two specific transporters for glycine, GlyT1 and GlyT2, perhaps with the cooperation of diverse neutral amino acid transporters like Asc-1 or SNAT5/SN2. While GlyT2 and Asc-1 are neuronal proteins, GlyT1 and SNAT5 are mainly astrocytic, although neuronal forms of GlyT1 also exist. GlyT1 has attracted considerable interest from the medical community and the pharmaceutical industry since compelling evidence indicates a clear association with the functioning of NMDARs, whose activity is decreased in various psychiatric illnesses. By controlling extracellular glycine, transporter inhibitors might potentiate the activity of NMDARs without activating excitotoxic processes. Physiologically, GlyT1 is a central actor in the cross talk between glutamatergic, glycinergic, dopaminergic, and probably other neurotransmitter systems. Many of these relationships begin to be unraveled by studies performed in recent years using genetic and pharmacological models. These studies are also clarifying the interactions between glycine, glycine transporters, and other co-agonists of the glycine site of NMDARs like D-serine. These findings are also relevant to understand the pathophysiology of devastating diseases like schizophrenia, depression, anxiety, epilepsy, stroke, and chronic pain.
Collapse
Affiliation(s)
- Francisco Zafra
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C / Nicolás Cabrera, 1, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras and IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain.
| | - Ignacio Ibáñez
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C / Nicolás Cabrera, 1, 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras and IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - David Bartolomé-Martín
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C / Nicolás Cabrera, 1, 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras and IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - Dolores Piniella
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C / Nicolás Cabrera, 1, 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras and IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - Marina Arribas-Blázquez
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C / Nicolás Cabrera, 1, 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras and IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - Cecilio Giménez
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C / Nicolás Cabrera, 1, 28049, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras and IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
48
|
Balu DT, Coyle JT. Altered CREB Binding to Activity-Dependent Genes in Serine Racemase Deficient Mice, a Mouse Model of Schizophrenia. ACS Chem Neurosci 2018; 9:2205-2209. [PMID: 29172439 DOI: 10.1021/acschemneuro.7b00404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
cAMP-response-element-binding protein (CREB) is a transcription factor ubiquitously expressed in the brain that regulates neuroplasticity by modulating gene expression. The influx of calcium through N-methyl-d-aspartate receptors (NMDARs) is a well-defined mechanism that leads to the increased expression of CREB-dependent genes, including brain derived neurotrophic factor (BDNF), microRNA-132, and activity-regulated cytoskeleton-associated protein (Arc). These molecules are implicated in the pathophysiology of schizophrenia. We previously demonstrated that serine racemase knockout (SR-/-) mice, which exhibit NMDAR hypofunction due to a lack of the forebrain NMDAR co-agonist d-serine, also have reduced expression of CREB-dependent genes in the hippocampus. Using chromatin immunoprecipitation, we show here that, in SR-/- mice, there is less CREB bound to the promoter regions of BDNF, microRNA-132, and Arc. These data suggest that NMDAR hypofunction in SR-/- mice leads to reduced CREB binding on known activity-dependent genes, in turn contributing to their reduced expression.
Collapse
Affiliation(s)
- Darrick T. Balu
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts 02115, United States
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, Massachusetts 02478, United States
| | - Joseph T. Coyle
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts 02115, United States
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, Massachusetts 02478, United States
| |
Collapse
|
49
|
D’Souza DC, Carson RE, Driesen N, Johannesen J, Ranganathan M, Krystal JH. Dose-Related Target Occupancy and Effects on Circuitry, Behavior, and Neuroplasticity of the Glycine Transporter-1 Inhibitor PF-03463275 in Healthy and Schizophrenia Subjects. Biol Psychiatry 2018; 84:413-421. [PMID: 29499855 PMCID: PMC6068006 DOI: 10.1016/j.biopsych.2017.12.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/14/2017] [Accepted: 12/15/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Glycine transporter-1 (GlyT1) inhibitors may ameliorate cognitive impairments associated with schizophrenia. The dose-related occupancy and target engagement of the GlyT1 inhibitor PF-03463275 were studied to inform optimal dose selection for a clinical trial for cognitive impairments associated with schizophrenia. METHODS In substudy 1, the effects of PF-03463275 (10, 20, and 40 mg twice a day) on occupancy of GlyT1 were tested using positron emission tomography and 18F-MK-6577, and visual long-term potentiation (LTP) in schizophrenia patients (SZs) and healthy control subjects. Furthermore, the capacity of PF-03463275 to attenuate ketamine-induced disruption of working memory-related activation of a "working memory" circuit was tested only in healthy control subjects using functional magnetic resonance imaging. Subsequently, the effects of PF-03463275 (60 mg twice a day) on occupancy of GlyT1 and long-term potentiation were examined only in SZs (substudy 2). RESULTS PF-03463275 at 10, 20, 40, and 60 mg twice a day produced ∼44%, 61%, 76%, and 83% GlyT1 occupancy, respectively, in SZs with higher ligand binding to GlyT1 in subcortical versus cortical regions. PF-03463275 did not attenuate any ketamine-induced effects but did improve working memory accuracy in healthy control subjects. PF-03463275 increased long-term potentiation only in SZs with peak effects at 40 mg twice a day (∼75% GlyT1 occupancy) and with a profile suggestive of an inverted U dose response. PF-03463275 was well-tolerated. CONCLUSIONS The dose-related GlyT1 occupancy of PF-03463275 is linear. While PF-03463275 did not show evidence of facilitating N-methyl-D-aspartate receptor function in the ketamine assay, it enhanced neuroplasticity in SZs. These findings provide support for a clinical trial to test the ability of PF-03463275 to enhance cognitive remediation toward addressing cognitive impairments associated with schizophrenia.
Collapse
Affiliation(s)
- Deepak Cyril D’Souza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Mental Health Service Line, VA Connecticut Healthcare System, West Haven, CT, USA,Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA
| | - Richard E. Carson
- Yale PET Center, New Haven, CT, USA,Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Naomi Driesen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Mental Health Service Line, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Jason Johannesen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Mental Health Service Line, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Mohini Ranganathan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Mental Health Service Line, VA Connecticut Healthcare System, West Haven, CT, USA,Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA
| | - John H. Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Mental Health Service Line, VA Connecticut Healthcare System, West Haven, CT, USA,Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA,Yale-New Haven Hospital, New Haven, CT,Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
50
|
Monitoring hippocampal glycine with the computationally designed optical sensor GlyFS. Nat Chem Biol 2018; 14:861-869. [DOI: 10.1038/s41589-018-0108-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 06/21/2018] [Indexed: 01/01/2023]
|