1
|
Konecny J, Misiachna A, Chvojkova M, Kleteckova L, Kolcheva M, Novak M, Prchal L, Ladislav M, Hemelikova K, Netolicky J, Hrabinova M, Kobrlova T, Karasova JZ, Pejchal J, Fibigar J, Vecera Z, Kucera T, Jendelova P, Zahumenska P, Langore E, Doderovic J, Pang YP, Vales K, Korabecny J, Soukup O, Horak M. Dizocilpine derivatives as neuroprotective NMDA receptor antagonists without psychomimetic side effects. Eur J Med Chem 2024; 280:116981. [PMID: 39442339 DOI: 10.1016/j.ejmech.2024.116981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
We aimed to prepare novel dibenzo [a,d][7]annulen derivatives that act on N-methyl-d-aspartate (NMDA) receptors with potential neuroprotective effects. Our approach involved modifying the tropane moiety of MK-801, a potent open-channel blocker known for its psychomimetic side effects, by introducing a seven-membered ring with substituted base moieties specifically to alleviate these undesirable effects. Our in silico analyses showed that these derivatives should have high gastrointestinal absorption and cross the blood-brain barrier (BBB). Our pharmacokinetic studies in rats supported this conclusion and confirmed the ability of leading compounds 3l and 6f to penetrate the BBB. Electrophysiological experiments showed that all compounds exhibited different inhibitory activity towards the two major NMDA receptor subtypes, GluN1/GluN2A and GluN1/GluN2B. Of the selected compounds intentionally differing in the inhibitory efficacy, 6f showed high relative inhibition (∼90 % for GluN1/GluN2A), while 3l showed moderate inhibition (∼50 %). An in vivo toxicity study determined that compounds 3l and 6f were safe at 10 mg/kg doses with no adverse effects. Behavioral studies demonstrated that these compounds did not induce hyperlocomotion or impair prepulse inhibition of startle response in rats. Neuroprotective assays using a model of NMDA-induced hippocampal neurodegeneration showed that compound 3l at a concentration of 30 μM significantly reduced hippocampal damage in rats. These results suggest that these novel dibenzo [a,d][7]annulen derivatives are promising candidates for developing NMDA receptor-targeted therapies with minimal psychotomimetic side effects.
Collapse
Affiliation(s)
- Jan Konecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Anna Misiachna
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Marketa Chvojkova
- National Institute of Mental Health, Topolova 748, 250 67, Klecany, Czech Republic
| | - Lenka Kleteckova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic; National Institute of Mental Health, Topolova 748, 250 67, Klecany, Czech Republic
| | - Marharyta Kolcheva
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Martin Novak
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Lukas Prchal
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Marek Ladislav
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Katarina Hemelikova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Jakub Netolicky
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Martina Hrabinova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Tereza Kobrlova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Jana Zdarova Karasova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jakub Fibigar
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Zbynek Vecera
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Tomas Kucera
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Pavla Jendelova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Petra Zahumenska
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Emily Langore
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Jovana Doderovic
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic
| | - Yuan-Ping Pang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, 55905, MN, USA
| | - Karel Vales
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic; National Institute of Mental Health, Topolova 748, 250 67, Klecany, Czech Republic
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic.
| | - Martin Horak
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 4, 14220, Prague, Czech Republic.
| |
Collapse
|
2
|
Misiachna A, Konecny J, Kolcheva M, Ladislav M, Prchal L, Netolicky J, Kortus S, Zahumenska P, Langore E, Novak M, Hemelikova K, Hermanova Z, Hrochova M, Pelikanova A, Odvarkova J, Pejchal J, Kassa J, Zdarova Karasova J, Korabecny J, Soukup O, Horak M. Potent and reversible open-channel blocker of NMDA receptor derived from dizocilpine with enhanced membrane-to-channel inhibition. Biomed Pharmacother 2024; 178:117201. [PMID: 39053419 DOI: 10.1016/j.biopha.2024.117201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) play a significant role in developing several central nervous system (CNS) disorders. Currently, memantine, used for treating Alzheimer's disease, and ketamine, known for its anesthetic and antidepressant properties, are two clinically used NMDAR open-channel blockers. However, despite extensive research into NMDAR modulators, many have shown either harmful side effects or inadequate effectiveness. For instance, dizocilpine (MK-801) is recognized for its powerful psychomimetic effects due to its high-affinity and nearly irreversible inhibition of the GluN1/GluN2 NMDAR subtypes. Unlike ketamine, memantine and MK-801 also act through a unique, low-affinity "membrane-to-channel inhibition" (MCI). We aimed to develop an open-channel blocker based on MK-801 with distinct inhibitory characteristics from memantine and MK-801. Our novel compound, K2060, demonstrated effective voltage-dependent inhibition in the micromolar range at key NMDAR subtypes, GluN1/GluN2A and GluN1/GluN2B, even in the presence of Mg2+. K2060 showed reversible inhibitory dynamics and a partially trapping open-channel blocking mechanism with a significantly stronger MCI than memantine. Using hippocampal slices, 30 µM K2060 inhibited excitatory postsynaptic currents in CA1 hippocampal neurons by ∼51 %, outperforming 30 µM memantine (∼21 % inhibition). K2060 exhibited No Observed Adverse Effect Level (NOAEL) of 15 mg/kg upon intraperitoneal administration in mice. Administering K2060 at a 10 mg/kg dosage resulted in brain concentrations of approximately 2 µM, with peak concentrations (Tmax) achieved within 15 minutes. Finally, applying K2060 with trimedoxime and atropine in mice exposed to tabun improved treatment outcomes. These results underscore K2060's potential as a therapeutic agent for CNS disorders linked to NMDAR dysfunction.
Collapse
Affiliation(s)
- Anna Misiachna
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Jan Konecny
- University Hospital Hradec Kralove, Biomedical Research Center, Sokolska 581, Hradec Kralove 500 05, Czech Republic; University of Defense, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, Hradec Kralove 50005, Czech Republic
| | - Marharyta Kolcheva
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Marek Ladislav
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Lukas Prchal
- University Hospital Hradec Kralove, Biomedical Research Center, Sokolska 581, Hradec Kralove 500 05, Czech Republic
| | - Jakub Netolicky
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Stepan Kortus
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Petra Zahumenska
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Emily Langore
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Martin Novak
- University Hospital Hradec Kralove, Biomedical Research Center, Sokolska 581, Hradec Kralove 500 05, Czech Republic
| | - Katarina Hemelikova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Zuzana Hermanova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Michaela Hrochova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Anezka Pelikanova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Jitka Odvarkova
- University Hospital Hradec Kralove, Biomedical Research Center, Sokolska 581, Hradec Kralove 500 05, Czech Republic; University of Defense, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, Hradec Kralove 50005, Czech Republic
| | - Jaroslav Pejchal
- University of Defense, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, Hradec Kralove 50005, Czech Republic
| | - Jiri Kassa
- University of Defense, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, Hradec Kralove 50005, Czech Republic
| | - Jana Zdarova Karasova
- University Hospital Hradec Kralove, Biomedical Research Center, Sokolska 581, Hradec Kralove 500 05, Czech Republic; University of Defense, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, Hradec Kralove 50005, Czech Republic
| | - Jan Korabecny
- University Hospital Hradec Kralove, Biomedical Research Center, Sokolska 581, Hradec Kralove 500 05, Czech Republic; University of Defense, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, Hradec Kralove 50005, Czech Republic
| | - Ondrej Soukup
- University Hospital Hradec Kralove, Biomedical Research Center, Sokolska 581, Hradec Kralove 500 05, Czech Republic; University of Defense, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, Hradec Kralove 50005, Czech Republic.
| | - Martin Horak
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic.
| |
Collapse
|
3
|
Boikov SI, Karelina TV, Sibarov DA, Antonov SM. Selective inhibitor of sodium-calcium exchanger, SEA0400, affects NMDA receptor currents and abolishes their calcium-dependent block by tricyclic antidepressants. Front Pharmacol 2024; 15:1432718. [PMID: 39156114 PMCID: PMC11327140 DOI: 10.3389/fphar.2024.1432718] [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: 05/16/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
The open-channel block of N-methyl-D-aspartate receptors (NMDARs) and their calcium-dependent desensitization (CDD) represent conventional mechanisms of glutamatergic synapse regulation. In neurotrauma, neurodegeneration, and neuropathic pain the clinical benefits of cure with memantine, ketamine, Mg2+, and some tricyclic antidepressants are often attributed to NMDAR open-channel block, while possible involvement of NMDAR CDD in the therapy is not well established. Here the effects of selective high-affinity sodium-calcium exchanger (NCX) isoform 1 inhibitor, SEA0400, on NMDA-activated whole-cell currents and their block by amitriptyline, desipramine and clomipramine recorded by patch-clamp technique in cortical neurons of primary culture were studied. We demonstrated that in the presence of extracellular Ca2+, 50 nM SEA0400 caused a reversible decrease of the steady-state amplitude of NMDAR currents, whereas loading neurons with BAPTA or the removal of extracellular Ca2+ abolished the effect. The decrease did not exceed 30% of the amplitude and did not depend on membrane voltage. The external Mg2+ block and 50 nM SEA0400 inhibition of currents were additive, suggesting their independent modes of action. In the presence of Ca2+ SEA0400 speeded up the decay of NMDAR currents to the steady state determined by CDD. The measured IC50 value of 27 nM for SEA0400-induced inhibition coincides with that for NCX1. Presumably, SEA0400 effects are induced by an enhancement of NMDAR CDD through the inhibition of Ca2+ extrusion by NCX1. SEA0400, in addition, at nanomolar concentrations could interfere with Ca2+-dependent effect of tricyclic antidepressants. In the presence of 50 nM SEA0400, the IC50s for NMDAR inhibition by amitriptyline and desipramine increased by about 20 folds, as the Ca2+-dependent NMDAR inhibition disappeared. This observation highlights NCX1 involvement in amitriptyline and desipramine effects on NMDARs and unmasks competitive relationships between SEA0400 and these antidepressants. Neither amitriptyline nor desipramine could affect NCX3. The open-channel block of NMDARs by these substances was not affected by SEA0400. In agreement, SEA0400 did not change the IC50 for clomipramine, which acts as a pure NMDAR open-channel blocker. Thus, NCX seems to represent a promising molecular target to treat neurological disorders, because of the ability to modulate NMDARs by decreasing the open probability through the enhancement of their CDD.
Collapse
Affiliation(s)
| | | | | | - Sergei M. Antonov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint-Petersburg, Russia
| |
Collapse
|
4
|
Tadros M, Rente Lavastida D, Hanna A. Therapeutic Potential of Intravenous Ketamine in Early-Onset Dementia: A Case Report. Cureus 2024; 16:e65261. [PMID: 39184713 PMCID: PMC11342591 DOI: 10.7759/cureus.65261] [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: 05/15/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
This case report discusses the use of intravenous (IV) ketamine as a potential therapeutic agent for early-onset dementia. A 56-year-old female with a diagnosis of early-onset dementia showed significant cognitive decline despite trying and failing several standard treatments such as memantine, donepezil, and rivastigmine. Given the promising results of ketamine in other neurological and psychiatric disorders, the patient underwent a series of IV ketamine infusions over a period of two months. Following treatment, there was a notable improvement in cognitive function, mood, and daily living activities. By the end of her treatments, the patient stated she had more mental clarity, increased focus, improved memory, and increased energy. This case highlights the potential use of ketamine as a novel treatment approach for early-onset dementia and warrants further investigation in larger clinical trials.
Collapse
Affiliation(s)
- Mariam Tadros
- College of Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, USA
| | | | - Ashraf Hanna
- Pain Management, Florida Spine Institute, Clearwater, USA
| |
Collapse
|
5
|
Stachowicz K, Pańczyszyn-Trzewik P, Misztak P, Rzeźniczek S, Sowa-Kućma M. Cyclooxygenase-2 inhibition affects the ratio of GluN2A/GluN2B receptor subunits through interaction with mGluR5 in the mouse brain. Neuropeptides 2024; 104:102409. [PMID: 38244260 DOI: 10.1016/j.npep.2024.102409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 01/22/2024]
Abstract
N-methyl-D-aspartic acid receptors (NMDARs) are the most studied receptors in mammalian brains. Their role in depression, cognition, schizophrenia, learning and memorization, Alzheimer's disease, and more is well documented. In the search for new drug candidates in depression, intensive studies have been conducted. Compounds that act by influencing NMDARs have been particularly intensively investigated following the success of ketamine in clinics. Unfortunately, the side effects associated with ketamine do not allow it to be useful in all cases. Therefore, it is important to learn about new unknown mechanisms related to NMDAR activation and study the impact of changes in the excitatory synapse environment on this receptor. Both direct and intermediary influence on NMDARs via mGluRs and COX-2 are effective. Our prior studies showed that both mGluRs ligands and COX-2 inhibitors are potent in depression-like and cognitive studies through mutual interactions. The side effects associated with imipramine administration, e.g., memory impairment, were improved when inhibiting COX-2. Therefore, this study is a trial that involves searching for modifications in NMDARs in mouse brains after prolonged treatment with MTEP (mGluR5 antagonist), NS398 (COX-2 inhibitor), or imipramine (tricyclic antidepressant). The prefrontal cortex (PFC) and hippocampus (HC) were selected for PCR and Western blot analyses. Altered expression of Gin2a or Grin2b genes after treatment was found. The observed effects were more potent when COX-2 was inhibited. The finding described here may be vital when searching for new drugs acting via NMDARs without the side effects related to cognition.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków 31-343, Poland.
| | - Patrycja Pańczyszyn-Trzewik
- Medical College of Rzeszów University, Institute of Medical Sciences, Department of Human Physiology, 35-310 Rzeszow, Kopisto Street 2a, Poland
| | - Paulina Misztak
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków 31-343, Poland
| | - Szymon Rzeźniczek
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków 31-343, Poland
| | - Magdalena Sowa-Kućma
- Medical College of Rzeszów University, Institute of Medical Sciences, Department of Human Physiology, 35-310 Rzeszow, Kopisto Street 2a, Poland
| |
Collapse
|
6
|
Karimi Tari P, Parsons CG, Collingridge GL, Rammes G. Memantine: Updating a rare success story in pro-cognitive therapeutics. Neuropharmacology 2024; 244:109737. [PMID: 37832633 DOI: 10.1016/j.neuropharm.2023.109737] [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: 02/16/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023]
Abstract
The great potential for NMDA receptor modulators as druggable targets in neurodegenerative disorders has been met with limited success. Considered one of the rare exceptions, memantine has consistently demonstrated restorative and prophylactic properties in many AD models. In clinical trials memantine slows the decline in cognitive performance associated with AD. Here, we provide an overview of the basic properties including pharmacological targets, toxicology and cellular effects of memantine. Evidence demonstrating reductions in molecular, physiological and behavioural indices of AD-like impairments associated with memantine treatment are also discussed. This represents both an extension and homage to Dr. Chris Parson's considerable contributions to our fundamental understanding of a success story in the AD treatment landscape.
Collapse
Affiliation(s)
- Parisa Karimi Tari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Chris G Parsons
- Galimedix Therapeutics, Inc., 2704 Calvend Lane, Kensington, 20895, MD, USA
| | - Graham L Collingridge
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada; Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada; TANZ Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care Medicine of the Technical University of Munich, School of Medicine, 22, 81675, Munich, Germany.
| |
Collapse
|
7
|
Iacobucci GJ, Popescu GK. Calcium- and calmodulin-dependent inhibition of NMDA receptor currents. Biophys J 2024; 123:277-293. [PMID: 38140727 PMCID: PMC10870176 DOI: 10.1016/j.bpj.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/05/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023] Open
Abstract
Calcium ions (Ca2+) reduce NMDA receptor currents through several distinct mechanisms. Among these, calmodulin (CaM)-dependent inhibition (CDI) accomplishes rapid, reversible, and incomplete reduction of the NMDA receptor currents in response to elevations in intracellular Ca2+. Quantitative and mechanistic descriptions of CDI of NMDA receptor-mediated signals have been marred by variability originating, in part, from differences in the conditions and metrics used to evaluate this process across laboratories. Recent ratiometric approaches to measure the magnitude and kinetics of NMDA receptor CDI have facilitated rapid insights into this phenomenon. Notably, the kinetics and magnitude of NMDA receptor CDI depend on the degree of saturation of its CaM binding sites, which represent the bona fide calcium sensor for this type of inhibition, the kinetics and magnitude of the Ca2+ signal, which depends on the biophysical properties of the NMDA receptor or of adjacent Ca2+ sources, and on the relative distribution of Ca2+ sources and CaM molecules. Given that all these factors vary widely during development, across cell types, and with physiological and pathological states, it is important to understand how NMDA receptor CDI develops and how it contributes to signaling in the central nervous system. Here, we review briefly these recent advances and highlight remaining questions about the structural and kinetic mechanisms of NMDA receptor CDI. Given that pathologies can arise from several sources, including mutations in the NMDA receptor and in CaM, understanding how CaM responds to intracellular Ca2+ signals to initiate conformational changes in NMDA receptors, and mapping the structural domains responsible will help to envision novel therapeutic strategies to neuropsychiatric diseases, which presently have limited available treatments.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, New York
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, New York.
| |
Collapse
|
8
|
Islam MS, Lai CC, Wang LH, Lin HH. Inhibition of NMDA Receptor Activation in the Rostral Ventrolateral Medulla by Amyloid-β Peptide in Rats. Biomolecules 2023; 13:1736. [PMID: 38136607 PMCID: PMC10741979 DOI: 10.3390/biom13121736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors, a subtype of ionotropic glutamate receptors, are important in regulating sympathetic tone and cardiovascular function in the rostral ventrolateral medulla (RVLM). Amyloid-beta peptide (Aβ) is linked to the pathogenesis of Alzheimer's disease (AD). Cerebro- and cardiovascular diseases might be the risk factors for developing AD. The present study examines the acute effects of soluble Aβ on the function of NMDA receptors in rats RVLM. We used the magnitude of increases in the blood pressure (pressor responses) induced by microinjection of NMDA into the RVLM as an index of NMDA receptor function in the RVLM. Soluble Aβ was applied by intracerebroventricular (ICV) injection. Aβ1-40 at a lower dose (0.2 nmol) caused a slight reduction, and a higher dose (2 nmol) showed a significant decrease in NMDA-induced pressor responses 10 min after administration. ICV injection of Aβ1-42 (2 nmol) did not affect NMDA-induced pressor responses in the RVLM. Co-administration of Aβ1-40 with ifenprodil or memantine blocked the inhibitory effects of Aβ1-40. Immunohistochemistry analysis showed a significant increase in the immunoreactivity of phosphoserine 1480 of GluN2B subunits (pGluN2B-serine1480) in the neuron of the RVLM without significant changes in phosphoserine 896 of GluN1 subunits (pGluN1-serine896), GluN1 and GluN2B, 10 min following Aβ1-40 administration compared with saline. Interestingly, we found a much higher level of Aβ1-40 compared to that of Aβ1-42 in the cerebrospinal fluid (CSF) measured using enzyme-linked immunosorbent assay 10 min following ICV administration of the same dose (2 nmol) of the peptides. In conclusion, the results suggest that ICV Aβ1-40, but not Aβ1-42, produced an inhibitory effect on NMDA receptor function in the RVLM, which might result from changes in pGluN2B-serine1480 (regulated by casein kinase II). The different elimination of the peptides in the CSF might contribute to the differential effects of Aβ1-40 and Aβ1-42 on NMDA receptor function.
Collapse
Affiliation(s)
- Md Sharyful Islam
- Master and Ph.D. Programs in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Chih-Chia Lai
- Department of Pharmacology, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Lan-Hui Wang
- Department of Physiology, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Hsun-Hsun Lin
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- Department of Physiology, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| |
Collapse
|
9
|
Liu W, Li Y, Zhao T, Gong M, Wang X, Zhang Y, Xu L, Li W, Li Y, Jia J. The role of N-methyl-D-aspartate glutamate receptors in Alzheimer's disease: From pathophysiology to therapeutic approaches. Prog Neurobiol 2023; 231:102534. [PMID: 37783430 DOI: 10.1016/j.pneurobio.2023.102534] [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: 02/27/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
N-Methyl-D-aspartate glutamate receptors (NMDARs) are involved in multiple physiopathological processes, including synaptic plasticity, neuronal network activities, excitotoxic events, and cognitive impairment. Abnormalities in NMDARs can initiate a cascade of pathological events, notably in Alzheimer's disease (AD) and even other neuropsychiatric disorders. The subunit composition of NMDARs is plastic, giving rise to a diverse array of receptor subtypes. While they are primarily found in neurons, NMDAR complexes, comprising both traditional and atypical subunits, are also present in non-neuronal cells, influencing the functions of various peripheral tissues. Furthermore, protein-protein interactions within NMDAR complexes has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation, and mitochondrial dysfunction, all of which potentially served as an obligatory relay of cognitive impairment. Nonetheless, the precise mechanistic link remains to be fully elucidated. In this review, we provided an in-depth analysis of the structure and function of NMDAR, investigated their interactions with various pathogenic proteins, discussed the current landscape of NMDAR-based therapeutics, and highlighted the remaining challenges during drug development.
Collapse
Affiliation(s)
- Wenying Liu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Tan Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Min Gong
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Xuechu Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yue Zhang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Wenwen Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China.
| |
Collapse
|
10
|
Milton AL. Drug memory reconsolidation: from molecular mechanisms to the clinical context. Transl Psychiatry 2023; 13:370. [PMID: 38040677 PMCID: PMC10692359 DOI: 10.1038/s41398-023-02666-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023] Open
Abstract
Since its rediscovery at the beginning of the 21st Century, memory reconsolidation has been proposed to be a therapeutic target for reducing the impact of emotional memories that can go awry in mental health disorders such as drug addiction (substance use disorder, SUD). Addiction can be conceptualised as a disorder of learning and memory, in which both pavlovian and instrumental learning systems become hijacked into supporting drug-seeking and drug-taking behaviours. The past two decades of research have characterised the details of the molecular pathways supporting the reconsolidation of pavlovian cue-drug memories, with more recent work indicating that the reconsolidation of instrumental drug-seeking memories also relies upon similar mechanisms. This narrative review considers what is known about the mechanisms underlying the reconsolidation of pavlovian and instrumental memories associated with drug use, how these approaches have translated to experimental medicine studies, and the challenges and opportunities for the clinical use of reconsolidation-based therapies.
Collapse
Affiliation(s)
- Amy L Milton
- Department of Psychology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Schmauder R, Eick T, Schulz E, Sammler G, Voigt E, Mayer G, Ginter H, Ditze G, Benndorf K. Fast functional mapping of ligand-gated ion channels. Commun Biol 2023; 6:1003. [PMID: 37783870 PMCID: PMC10545696 DOI: 10.1038/s42003-023-05340-w] [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: 04/06/2023] [Accepted: 09/11/2023] [Indexed: 10/04/2023] Open
Abstract
Ligand-gated ion channels are formed by three to five subunits that control the opening of the pore in a cooperative fashion. We developed a microfluidic chip-based technique for studying ion currents and fluorescence signals in either excised membrane patches or whole cells to measure activation and deactivation kinetics of the channels as well as ligand binding and unbinding when using confocal patch-clamp fluorometry. We show how this approach produces in a few seconds either unidirectional concentration-activation relationships at or near equilibrium and, moreover, respective time courses of activation and deactivation for a large number of freely designed steps of the ligand concentration. The short measuring period strongly minimizes the contribution of disturbing superimposing effects such as run-down phenomena and desensitization effects. To validate gating mechanisms, complex kinetic schemes are quantified without the requirement to have data at equilibrium. The new method has potential for functionally analyzing any ligand-gated ion channel and, beyond, also for other receptors.
Collapse
Affiliation(s)
- Ralf Schmauder
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07743, Jena, Germany.
| | - Thomas Eick
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07743, Jena, Germany
| | - Eckhard Schulz
- Hochschule Schmalkalden, Fakultät Elektrotechnik, Blechhammer, 98574, Schmalkalden, Germany
| | - Günther Sammler
- Zentrale Forschungswerkstätten, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07743, Jena, Germany
| | - Elmar Voigt
- Leibniz Institut für Photonische Technologien e.V., Albert-Einstein-Straße 9, 07745, Jena, Germany
| | - Günter Mayer
- Leibniz Institut für Photonische Technologien e.V., Albert-Einstein-Straße 9, 07745, Jena, Germany
| | - Holger Ginter
- Zentrale Forschungswerkstätten, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07743, Jena, Germany
| | - Günter Ditze
- Zentrale Forschungswerkstätten, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07743, Jena, Germany
| | - Klaus Benndorf
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07743, Jena, Germany.
| |
Collapse
|
12
|
McClure EW, Daniels RN. Classics in Chemical Neuroscience: Dextromethorphan (DXM). ACS Chem Neurosci 2023. [PMID: 37290117 DOI: 10.1021/acschemneuro.3c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
Dextromethorphan (DXM) was introduced in 1958 as the first non-opioid cough suppressant and is indicated for multiple psychiatric disorders. It has been the most used over-the-counter cough suppressant since its emergence. However, individuals quickly noticed an intoxicating and psychedelic effect if they ingested large doses. DXM's antagonism at N-methyl-d-aspartate receptors (NMDAr) is thought to underly its efficacy in treating acute cough, but supratherapeutic doses mimic the activity of dissociative hallucinogens, such as phencyclidine and ketamine. In this Review we will discuss DXM's synthesis, manufacturing information, drug metabolism, pharmacology, adverse effects, recreational use, abuse potential, and its history and importance in therapy to present DXM as a true classic in chemical neuroscience.
Collapse
Affiliation(s)
- Elliot W McClure
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, Tennessee 37204, United States
| | - R Nathan Daniels
- Department of Pharmaceutical Sciences, Union University College of Pharmacy, Jackson, Tennessee 38305, United States
| |
Collapse
|
13
|
Ferguson AA, Khan AI, Abuzainah B, Chaudhuri D, Khan KI, Al Shouli R, Allakky A, Hamdan JA. Clinical Effectiveness of N-Methyl-D-Aspartate (NMDA) Receptor Antagonists in Adult Obsessive-Compulsive Disorder (OCD) Treatment: A Systematic Review. Cureus 2023; 15:e37833. [PMID: 37213965 PMCID: PMC10198239 DOI: 10.7759/cureus.37833] [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: 10/27/2022] [Accepted: 04/13/2023] [Indexed: 05/23/2023] Open
Abstract
Obsessive-compulsive disorder (OCD) is a neuropsychiatric disorder that affects approximately 2% of the human population. Traditional treatment of OCD includes selective serotonin reuptake inhibitor (SSRI) or serotonin reuptake inhibitor (SRI) treatment along with cognitive behavioral therapy (CBT). Nearly 25%-30% of OCD patients do not respond to SSRIs. Glutamatergic agents are currently being studied for the treatment of OCD due to the glutamatergic pathway in the brain, related to OCD, and the role of the cortico-striato-thalamic circuit (CSTC). This review assesses the clinical effectiveness of N-methyl-D-aspartate (NMDA) antagonists, ketamine/esketamine, memantine, and amantadine, for adult patients with OCD. Inclusion criteria include human studies published within the last 15 years, with patients diagnosed with OCD, aged over 18 years, with only psychiatric comorbidities, and full-text articles. Papers that included interventions other than CBT, exposure with response prevention (ERP), and SSRI/SRI were excluded. Articles were searched for using PubMed, PubMed Central, Medical Literature Analysis and Retrieval System Online, GeorgiA LIbrary LEarning Online, EBSCO Information Services, OpenAthens, Multidisciplinary Digital Publishing Institute, and Google Scholar databases, last searched on December 2, 2022. The risk of bias was assessed using Cochrane Risk of Bias tools, the Scale for the Assessment of Narrative Review Articles (SANRA) checklist for literature reviews, and the Joanna Briggs Institute (JBI) Critical Appraisal Checklist for quasi-experimental studies. Results were presented and synthesized by Excel spreadsheet analysis. The database search yielded 4,221 articles, which was cut down to 18 articles by inclusion/exclusion criteria, including duplications. 80% of the ketamine studies resulted in a significant reduction of obsessions and compulsions based on the Yale-Brown Obsessive-Compulsive Scale (Y-BOCS), and each of the memantine and amantadine studies displayed clinical effectiveness, also. Limitations include the small number of amantadine studies and the limited availability of other NMDA receptor (NMDAR) antagonist-focused studies. This systematic review shows that ketamine is an effective drug for the treatment of non-refractory, mild to moderate OCD, and memantine and amantadine are effective augmentation agents for the treatment of mild to severe OCD.
Collapse
Affiliation(s)
- Asila A Ferguson
- Psychiatry, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aujala Irfan Khan
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Baraa Abuzainah
- General Practice, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Dipabali Chaudhuri
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Kokab Irfan Khan
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Roba Al Shouli
- Pediatric, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Akhil Allakky
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jaafar A Hamdan
- Medicine, American University of Antigua, St. John, ATG
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
14
|
Iacobucci GJ, Liu B, Wen H, Sincox B, Zheng W, Popescu GK. Complex functional phenotypes of NMDA receptor disease variants. Mol Psychiatry 2022; 27:5113-5123. [PMID: 36117210 DOI: 10.1038/s41380-022-01774-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/23/2022] [Accepted: 08/31/2022] [Indexed: 01/14/2023]
Abstract
NMDA receptors have essential roles in the physiology of central excitatory synapses and their dysfunction causes severe neuropsychiatric symptoms. Recently, a series of genetic variants have been identified in patients, however, functional information about these variants is sparse and their role in pathogenesis insufficiently known. Here we investigate the mechanism by which two GluN2A variants may be pathogenic. We use molecular dynamics simulation and single-molecule electrophysiology to examine the contribution of GluN2A subunit-residues, P552 and F652, and their pathogenic substitutions, P552R and F652V, affect receptor functions. We found that P552 and F652 interact during the receptors' normal activity cycle; the interaction stabilizes receptors in open conformations and is required for a normal electrical response. Engineering shorter side-chains at these positions (P552A and/or F652V) caused a loss of interaction energy and produced receptors with severe gating, conductance, and permeability deficits. In contrast, the P552R side chain resulted in stronger interaction and produced a distinct, yet still drastically abnormal electrical response. These results identify the dynamic contact between P552 and F652 as a critical step in the NMDA receptor activation, and show that both increased and reduced communication through this interaction cause dysfunction. Results show that subtle differences in NMDA receptor primary structure can generate complex phenotypic alterations whose binary classification is too simplistic to serve as a therapeutic guide.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Beiying Liu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Han Wen
- Department of Physics, College of Arts and Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Brittany Sincox
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Wenjun Zheng
- Department of Physics, College of Arts and Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
15
|
Ornowska M, Wormsbecker A, Andolfatto G, Leung T, Khan I, Medvedev G. The use of ketamine as a neuroprotective agent following cardiac arrest: A scoping review of current literature. CNS Neurosci Ther 2022; 29:104-110. [PMID: 36184822 PMCID: PMC9804040 DOI: 10.1111/cns.13983] [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: 04/05/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 02/06/2023] Open
Abstract
AIMS The objective of this article is to summarize the state of the literature surrounding the use of ketamine as a neuroprotective agent following cardiac arrest. METHODS Five electronic databases were used to search for studies related to the use of ketamine for neuroprotection following cardiac arrest. This search was performed once in May 2020, and an updated search was conducted in May 2021 and March 2022. RESULTS All searches combined retrieved 181 results; no clinical trials were identified. As such, the authors were limited to writing a scoping review of the literature rather than a systematic review. CONCLUSIONS The current state of the literature describes the mechanism of action of ketamine as a neuroprotective agent through its action as an NMDA antagonist. There is evidence of its efficacy as a neuroprotective agent in preclinical models of cardiac arrest. Current published clinical work supports the use of ketamine ameliorating neurologic outcomes in other conditions such as epilepsy, traumatic brain injury, and depression. The current state of the literature is reflective of the notion that the use of ketamine following cardiac arrest may result in improved neurologic outcomes. Future research directions should focus on the use of ketamine as a possible clinical intervention following cardiac arrest.
Collapse
Affiliation(s)
- Marlena Ornowska
- Department of Biomedical PhysiologySimon Fraser UniversityBurnabyBritish ColumbiaCanada
| | - Andrew Wormsbecker
- Royal Columbian Hospital, Fraser Health AuthorityNew WestminsterBritish ColumbiaCanada,Division of Critical Care, Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Gary Andolfatto
- Department of Emergency Medicine, Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Tim S. Leung
- Royal Columbian Hospital, Fraser Health AuthorityNew WestminsterBritish ColumbiaCanada,Faculty of Pharmaceutical SciencesUniversity of British ColumbiaVancouverBritish ColumbiaCanada,Lower Mainland Pharmacy ServicesVancouverBritish ColumbiaCanada
| | - Idan Khan
- Royal Columbian Hospital, Fraser Health AuthorityNew WestminsterBritish ColumbiaCanada,Department of Emergency Medicine, Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - George Medvedev
- Royal Columbian Hospital, Fraser Health AuthorityNew WestminsterBritish ColumbiaCanada,Division of Neurology, Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
16
|
da Silva Beggiora P, da Silva SC, Rodrigues KP, Almeida TADL, Botelho GS, Silva GAPDM, Machado HR, da Silva Lopes L. Memantine associated with ventricular-subcutaneous shunt promotes behavioral improvement, reduces reactive astrogliosis and cell death in juvenile hydrocephalic rats. J Chem Neuroanat 2022; 125:102165. [PMID: 36152798 DOI: 10.1016/j.jchemneu.2022.102165] [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: 06/29/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/28/2022]
Abstract
Hydrocephalus is defined as the accumulation of cerebrospinal fluid in the brain ventricles. The usual treatment of hydrocephalus is surgical (shunt), but not all patients can undergo treatment immediately after diagnosis. Thus, neuroprotective measures were tested to minimize the tissue damage involved. Memantine is a non-competitive antagonist of the N-methyl-D-aspartate (NMDA) receptor, which has shown a neuroprotective action in neurodegenerative diseases. This study aimed to evaluate the neuroprotective response of memantine in animals treated with or without a ventricular-subcutaneous shunt. Seven-day-old male Wistar rats induced by intracisternal injection of kaolin were used, divided into five groups: intact control (n=10), hydrocephalic (n=10), hydrocephalic treated with memantine (20mg/kg/day) (n=10), hydrocephalic treated with shunt (n=10), hydrocephalic treated with shunt and memantine (20mg/kg/day) (n=10). Memantine administration was started on the day after hydrocephalus induction and continued until the last day of the experimental period, totaling 21 consecutive days of drug application. The CSF shunt surgery was performed seven days after hydrocephalus induction. Behavioral tests (open field, and modified Morris water maze), histological, and immunohistochemical evaluations were performed. Treatment with memantine resulted in significant improvement (p<0.05) in sensorimotor development, preservation of spatial memory, reduction of astrocytic reaction in the corpus callosum, cortex, and germinal matrix. When associated with the shunt, it has also been shown to reduce the cell death cascade. It is concluded that memantine is a promising adjuvant drug with beneficial potential for the treatment of lesions secondary to hydrocephalus.
Collapse
Affiliation(s)
- Pâmella da Silva Beggiora
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Av, 3900, Ribeirão Preto, SP, 14040-900, Brazil.
| | - Stephanya Covas da Silva
- Department of Morphology and Pathology, Federal University of São Carlos, Washington Luiz, Monjolinho, São Carlos - SP, 13565-905, Brazil.
| | - Karine Pereira Rodrigues
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Av, 3900, Ribeirão Preto, SP, 14040-900, Brazil.
| | - Timóteo Abrantes de Lacerda Almeida
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Av, 3900, Ribeirão Preto, SP, 14040-900, Brazil.
| | - Gustavo Sampaio Botelho
- Department of Pediatric Neurosurgery, Children's and Maternity Hospital of São José do Rio Preto. Jamil Ferreira Kfouri Av, 3355, São José do Rio Preto, SP, 15091-240, Brazil.
| | - Gabriel Aparecido Pinto de Moura Silva
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Av, 3900, Ribeirão Preto, SP, 14040-900, Brazil.
| | - Hélio Rubens Machado
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Av, 3900, Ribeirão Preto, SP, 14040-900, Brazil.
| | - Luiza da Silva Lopes
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Av, 3900, Ribeirão Preto, SP, 14040-900, Brazil.
| |
Collapse
|
17
|
Inhibition of NMDA receptors through a membrane-to-channel path. Nat Commun 2022; 13:4114. [PMID: 35840593 PMCID: PMC9287434 DOI: 10.1038/s41467-022-31817-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 07/05/2022] [Indexed: 11/21/2022] Open
Abstract
N-methyl-d-aspartate receptors (NMDARs) are transmembrane proteins that are activated by the neurotransmitter glutamate and are found at most excitatory vertebrate synapses. NMDAR channel blockers, an antagonist class of broad pharmacological and clinical significance, inhibit by occluding the NMDAR ion channel. A vast literature demonstrates that NMDAR channel blockers, including MK-801, phencyclidine, ketamine, and the Alzheimer’s disease drug memantine, can bind and unbind only when the NMDAR channel is open. Here we use electrophysiological recordings from transfected tsA201 cells and cultured neurons, NMDAR structural modeling, and custom-synthesized compounds to show that NMDAR channel blockers can enter the channel through two routes: the well-known hydrophilic path from extracellular solution to channel through the open channel gate, and also a hydrophobic path from plasma membrane to channel through a gated fenestration (“membrane-to-channel inhibition” (MCI)). Our demonstration that ligand-gated channels are subject to MCI, as are voltage-gated channels, highlights the broad expression of this inhibitory mechanism. Wilcox et al. (2022) show that NMDA receptor channel blockers, some of which are clinically important drugs, can access their binding site via 2 routes: a well-known path from the extracellular solution, and another path through the plasma membrane.
Collapse
|
18
|
Maeda S, Yamada J, Iinuma KM, Nadanaka S, Kitagawa H, Jinno S. Chondroitin sulfate proteoglycan is a potential target of memantine to improve cognitive function via the promotion of adult neurogenesis. Br J Pharmacol 2022; 179:4857-4877. [PMID: 35797426 DOI: 10.1111/bph.15920] [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: 10/27/2021] [Revised: 06/02/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Chondroitin sulfate proteoglycan (CSPG) constitutes the neurogenic niche in the hippocampus. The reduction of hippocampal neurogenesis is involved in aging-related cognitive decline and dementia. The purpose of this study is to find candidates that improve cognitive function by analyzing the effects of memantine (MEM), a therapeutic agent for Alzheimer's disease, on CSPG and adult hippocampal neurogenesis. EXPERIMENTAL APPROACH The effects of MEM on neurogenesis-related cells and CSPG content were assessed in the hippocampus of middle-aged mice. The MEM-induced alterations in gene expressions of neurotrophins and enzymes associated with biosynthesis and degradation of CSPG in the hippocampus were also measured. The effects of MEM on cognitive function were estimated using a behavioral test battery. The same set of behavioral tests was applied to evaluate the effects of pharmacological depletion of CSPG in the hippocampus. KEY RESULTS The densities of newborn granule cells and content of CSPG in the hippocampus were increased by MEM. The expression levels of the enzyme responsible for the biosynthesis CSPG were increased by MEM. The neurotrophin-related molecules were activated by MEM. Short- and long-term memory performance was improved by MEM. Pharmacological depletion of CSPG impairs the effects of MEM on cognitive improvement in middle-aged mice. CONCLUSIONS AND IMPLICATIONS MEM regulates the biosynthesis and degradation of CSPG, which may underlie the improvement of cognitive function via the promotion of adult hippocampal neurogenesis. These results imply that CSPG-related enzymes may be potentially attractive candidates for the treatment of aging-related cognitive decline.
Collapse
Affiliation(s)
- Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satomi Nadanaka
- Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Hiroshi Kitagawa
- Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
19
|
Xu S, Yao X, Li B, Cui R, Zhu C, Wang Y, Yang W. Uncovering the Underlying Mechanisms of Ketamine as a Novel Antidepressant. Front Pharmacol 2022; 12:740996. [PMID: 35872836 PMCID: PMC9301111 DOI: 10.3389/fphar.2021.740996] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
Major depressive disorder (MDD) is a devastating psychiatric disorder which exacts enormous personal and social-economic burdens. Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, has been discovered to exert rapid and sustained antidepressant-like actions on MDD patients and animal models. However, the dissociation and psychotomimetic propensities of ketamine have limited its use for psychiatric indications. Here, we review recently proposed mechanistic hypotheses regarding how ketamine exerts antidepressant-like actions. Ketamine may potentiate α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR)-mediated transmission in pyramidal neurons by disinhibition and/or blockade of spontaneous NMDAR-mediated neurotransmission. Ketamine may also activate neuroplasticity- and synaptogenesis-relevant signaling pathways, which may converge on key components like brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) and mechanistic target of rapamycin (mTOR). These processes may subsequently rebalance the excitatory/inhibitory transmission and restore neural network integrity that is compromised in depression. Understanding the mechanisms underpinning ketamine’s antidepressant-like actions at cellular and neural circuit level will drive the development of safe and effective pharmacological interventions for the treatment of MDD.
Collapse
Affiliation(s)
- Songbai Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Cuilin Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| | - Yao Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| |
Collapse
|
20
|
Bartsch CJ, Nordman JC. Promises and Pitfalls of NMDA Receptor Antagonists in Treating Violent Aggression. Front Behav Neurosci 2022; 16:938044. [PMID: 35801096 PMCID: PMC9253591 DOI: 10.3389/fnbeh.2022.938044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Treatment options for chronically aggressive individuals remain limited despite recent medical advances. Traditional pharmacological agents used to treat aggression, such as atypical antipsychotics, have limited efficacy and are often replete with dangerous side effects. The non-competitive NMDAR antagonists ketamine and memantine are promising alternatives, but their effects appear to be highly dependent on dosage, context, and personal experience. Importantly, these drugs can increase aggression when combined with substances of abuse or during periods of heightened stress. This is likely due to mechanistic differences operating at specific synapses under different contexts. Previous findings from our lab and others have shown that early life stress, substance abuse, and attack experience promote aggression through NMDAR-dependent synaptic plasticity within aggression-related brain circuits. Ketamine and memantine affect these types of aggression in opposite ways. This has led us to propose that ketamine and memantine oppositely affect aggression brought on by early life stress, substance abuse, or attack experience through opposite effects on NMDAR-dependent synaptic plasticity. This would account for the persistent effects of these drugs on aggression and suggest they could be leveraged as a more long-lasting treatment option. However, a more thorough examination of the effects of ketamine and memantine on cellular and synaptic function will be necessary for responsible administration. Additionally, because the effects of ketamine and memantine are highly dependent on prior drug use, traumatic stress, or a history of aggressive behavior, we propose a more thorough medical evaluation and psychiatric assessment will be necessary to avoid possible adverse interactions with these drugs.
Collapse
Affiliation(s)
- Caitlyn J. Bartsch
- Department of Physiology, University of Southern Illinois Carbondale, Carbondale, IL, United States
| | - Jacob C. Nordman
- Department of Physiology, University of Southern Illinois School of Medicine, Carbondale, IL, United States
- *Correspondence: Jacob C. Nordman
| |
Collapse
|
21
|
Structural insights into binding of therapeutic channel blockers in NMDA receptors. Nat Struct Mol Biol 2022; 29:507-518. [PMID: 35637422 PMCID: PMC10075384 DOI: 10.1038/s41594-022-00772-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/04/2022] [Indexed: 01/02/2023]
Abstract
Excitatory signaling mediated by N-methyl-D-aspartate receptor (NMDAR) is critical for brain development and function, as well as for neurological diseases and disorders. Channel blockers of NMDARs are of medical interest owing to their potential for treating depression, Alzheimer's disease, and epilepsy. However, precise mechanisms underlying binding and channel blockade have remained limited owing to challenges in obtaining high-resolution structures at the binding site within the transmembrane domains. Here, we monitor the binding of three clinically important channel blockers: phencyclidine, ketamine, and memantine in GluN1-2B NMDARs at local resolutions of 2.5-3.5 Å around the binding site using single-particle electron cryo-microscopy, molecular dynamics simulations, and electrophysiology. The channel blockers form different extents of interactions with the pore-lining residues, which control mostly off-speeds but not on-speeds. Our comparative analyses of the three unique NMDAR channel blockers provide a blueprint for developing therapeutic compounds with minimal side effects.
Collapse
|
22
|
Fan H, Tong Z, Ren Z, Mishra K, Morita S, Edouarzin E, Gorla L, Averkiev B, Day VW, Hua DH. Synthesis and Characterization of Bimetallic Nanoclusters Stabilized by Chiral and Achiral Polyvinylpyrrolidinones. Catalytic C(sp 3)-H Oxidation. J Org Chem 2022; 87:6742-6759. [PMID: 35511477 DOI: 10.1021/acs.joc.2c00449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Second-generation chiral-substituted poly-N-vinylpyrrolidinones (CSPVPs) (-)-1R and (+)-1S were synthesized by free-radical polymerization of (3aR,6aR)- and (3aS,6aS)-5-ethenyl-tetrahydro-2,2-dimethyl-4H-1,3-dioxolo[4,5-c]pyrrol-4-one, respectively, using thermal and photochemical reactions. They were produced from respective d-isoascorbic acid and d-ribose. In addition, chiral polymer (-)-2 was also synthesized from the polymerization of (S)-3-(methoxymethoxy)-1-vinylpyrrolidin-2-one. Molecular weights of these chiral polymers were measured using HRMS, and the polymer chain tacticity was studied using 13C NMR spectroscopy. Chiral polymers (-)-1R, (+)-1S, and (-)-2 along with poly-N-vinylpyrrolidinone (PVP, MW 40K) were separately used in the stabilization of Cu/Au or Pd/Au nanoclusters. CD spectra of the bimetallic nanoclusters stabilized by (-)-1R and (+)-1S showed close to mirror-imaged CD absorption bands at wavelengths 200-300 nm, revealing that bimetallic nanoclusters' chiroptical responses are derived from chiral polymer-encapsulated nanomaterials. Chemo-, regio-, and stereo-selectivity was found in the catalytic C-H group oxidation reactions of complex bioactive natural products, such as ambroxide, menthofuran, boldine, estrone, dehydroabietylamine, 9-allogibberic acid, and sclareolide, and substituted adamantane molecules, when catalyst Cu/Au (3:1) or Pd/Au (3:1) stabilized by CSPVPs or PVP and oxidant H2O2 or t-BuOOH were applied. Oxidation of (+)-boldine N-oxide 23 using NMO as an oxidant yielded 4,5-dehydroboldine 27, and oxidation of (-)-9-allogibberic acid yielded C6,15 lactone 47 and C6-ketone 48.
Collapse
Affiliation(s)
- Huafang Fan
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Zongbo Tong
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Zhaoyang Ren
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Kanchan Mishra
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Shunya Morita
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Edruce Edouarzin
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Lingaraju Gorla
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Boris Averkiev
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Victor W Day
- Department of Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Duy H Hua
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| |
Collapse
|
23
|
Stepanenko YD, Sibarov DA, Shestakova NN, Antonov SM. Tricyclic Antidepressant Structure-Related Alterations in Calcium-Dependent Inhibition and Open-Channel Block of NMDA Receptors. Front Pharmacol 2022; 12:815368. [PMID: 35237149 PMCID: PMC8882908 DOI: 10.3389/fphar.2021.815368] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/27/2021] [Indexed: 12/30/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are an essential target for the analgetic action of tricyclic antidepressants (TCAs). Their therapeutic blood concentrations achieve 0.5–1.5 μM, which, however, are insufficient to cause in vitro the open-channel block known as the only effect of TCAs on NMDARs. Whereas structures of amitriptyline (ATL), desipramine (DES), and clomipramine (CLO) are rather similar these compounds manifest different therapeutic profiles and side effects. To study structure-activity relationships of DES and CLO on NMDARs, we measured IC50s as a function of extracellular calcium ([Ca2+]) and membrane voltage (Vm) of NMDAR currents recorded in cortical neurons. Here two components of TCA action on NMDARs are described, which could be characterized as the Ca2+-dependent inhibition and the open-channel block. DES demonstrated a profound Ca2+-dependent inhibition of NMDARs, while the CLO effect was weak. DES IC50 exhibited an e-fold change with a [Ca2+] shift of 0.59 mM, which is consistent with ATL. The Ca2+ dependence of NMDAR inhibition by DES disappeared in BAPTA loaded neurons, suggesting that Ca2+ acts from the inside. Since CLO differs from DES and ATL by the presence of Cl-atom in the structure, most likely, this is the atom which is responsible for the loss of pronounced [Ca2+] dependence. As for the NMDAR open-channel block, both DES and CLO were about 5-folds more potent than ATL due to their slow rates of dissociation either from open and closed states. DES demonstrated stronger Vm-dependence than CLO, suggesting a deeper location of the DES binding site within the ion pore. Because DES and CLO differ from ATL by the nitrogen-containing tricycle, presumably this moiety of the molecules determines their high-affinity binding with the NMDAR channel, while the aliphatic chain mono-methyl amino-group of DES allows a deep permeation in the channel. Thus, different structure-activity relationships of the Ca2+-dependent inhibition and Vm-dependent open-channel block of NMDARs by DES and CLO suggest that these processes are independent and most likely may represent an action on different molecular targets. The proposed model of TCA action on NMDARs predicts well the experimental values of IC50s at physiological [Ca2+] and within a wide range of Vms.
Collapse
|
24
|
Morningstar MD, Barnett WH, Goodlett CR, Kuznetsov A, Lapish CC. Understanding ethanol's acute effects on medial prefrontal cortex neural activity using state-space approaches. Neuropharmacology 2021; 198:108780. [PMID: 34480911 PMCID: PMC8488975 DOI: 10.1016/j.neuropharm.2021.108780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/10/2021] [Accepted: 08/30/2021] [Indexed: 12/22/2022]
Abstract
Acute ethanol (EtOH) intoxication results in several maladaptive behaviors that may be attributable, in part, to the effects of EtOH on neural activity in medial prefrontal cortex (mPFC). The acute effects of EtOH on mPFC function have been largely described as inhibitory. However, translating these observations on function into a mechanism capable of delineating acute EtOH's effects on behavior has proven difficult. This review highlights the role of acute EtOH on electrophysiological measurements of mPFC function and proposes that interpreting these changes through the lens of dynamical systems theory is critical to understand the mechanisms that mediate the effects of EtOH intoxication on behavior. Specifically, the present review posits that the effects of EtOH on mPFC N-methyl-d-aspartate (NMDA) receptors are critical for the expression of impaired behavior following EtOH consumption. This hypothesis is based on the observation that recurrent activity in cortical networks is supported by NMDA receptors, and, when disrupted, may lead to impairments in cognitive function. To evaluate this hypothesis, we discuss the representation of mPFC neural activity in low-dimensional, dynamic state spaces. This approach has proven useful for identifying the underlying computations necessary for the production of behavior. Ultimately, we hypothesize that EtOH-related alterations to NMDA receptor function produces alterations that can be effectively conceptualized as impairments in attractor dynamics and provides insight into how acute EtOH disrupts forms of cognition that rely on mPFC function. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.
Collapse
Affiliation(s)
| | - William H Barnett
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA
| | - Charles R Goodlett
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| | - Alexey Kuznetsov
- Indiana University-Purdue University Indianapolis, Department of Mathematics, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| | - Christopher C Lapish
- Indiana University-Purdue University Indianapolis, Department of Psychology, USA; Indiana University School of Medicine, Stark Neurosciences, USA
| |
Collapse
|
25
|
Nikolaev MV, Strashkov DM, Ryazantsev MN, Tikhonov DB. Optical Control of N-Methyl-d-aspartate Receptors by Azobenzene Quaternary Ammonium Compounds. ACS Chem Neurosci 2021; 12:3347-3357. [PMID: 34469111 DOI: 10.1021/acschemneuro.1c00310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Azobenzene-based quaternary ammonium compounds provide optical control of ion channels and are considered promising agents for regulation of neuronal excitability and for restoration of the photosensitivity of retinal cells. However, the selectivity of the action of these compounds remains insufficiently known. We studied the action of DENAQ (diethylamine-azobenzene-quaternary ammonium) and DMNAQ (dimethylamine-azobenzene-quaternary ammonium) on ionotropic glutamate receptors in rat brain neurons. In the dark, both compounds applied extracellularly caused fast and reversible inhibition of NMDA (N-methyl-d-aspartate) receptor-mediated currents with IC50 values of 10 and 5 μM, respectively. Light-induced transformation of DENAQ and DMNAQ to their cis forms caused the IC50 values to increase to 30 and 27 μM, respectively. Detailed analysis of this action revealed a complex nature consisting of fast inhibitory and slower potentiating effects. The AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors were only weakly affected independently on illumination. We conclude that, in addition to their long-lasting intracellular action, which persists after washout, azobenzene-based quaternary ammonium compounds should affect glutamatergic transmission and synaptic plasticity during treatment. Our findings also extend the list of soluble photoswitchable inhibitors of NMDA receptors. While the site(s) and mechanisms of action are unclear, the effect of DENAQ demonstrates strong pH dependence. At acidic pH values, DENAQ potentiates both NMDA and AMPA receptors.
Collapse
Affiliation(s)
- Maxim V. Nikolaev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, Thorez pr. 44, 194223 Saint Petersburg, Russia
| | - Daniil M. Strashkov
- Saint Petersburg National Research Academic University of the Russian Academy of Sciences, 8/3 Khlopina Street, 194021 Saint Petersburg, Russia
| | - Mikhail N. Ryazantsev
- Institute of Chemistry, Saint Petersburg State University, Universitetskii pr. 26, 198504 Saint Petersburg, Russia
| | - Denis B. Tikhonov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, Thorez pr. 44, 194223 Saint Petersburg, Russia
| |
Collapse
|
26
|
High-frequency head impact causes chronic synaptic adaptation and long-term cognitive impairment in mice. Nat Commun 2021; 12:2613. [PMID: 33972519 PMCID: PMC8110563 DOI: 10.1038/s41467-021-22744-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 03/24/2021] [Indexed: 02/03/2023] Open
Abstract
Repeated head impact exposure can cause memory and behavioral impairments. Here, we report that exposure to non-damaging, but high frequency, head impacts can alter brain function in mice through synaptic adaptation. High frequency head impact mice develop chronic cognitive impairments in the absence of traditional brain trauma pathology, and transcriptomic profiling of mouse and human chronic traumatic encephalopathy brain reveal that synapses are strongly affected by head impact. Electrophysiological analysis shows that high frequency head impacts cause chronic modification of the AMPA/NMDA ratio in neurons that underlie the changes to cognition. To demonstrate that synaptic adaptation is caused by head impact-induced glutamate release, we pretreated mice with memantine prior to head impact. Memantine prevents the development of the key transcriptomic and electrophysiological signatures of high frequency head impact, and averts cognitive dysfunction. These data reveal synapses as a target of high frequency head impact in human and mouse brain, and that this physiological adaptation in response to head impact is sufficient to induce chronic cognitive impairment in mice.
Collapse
|
27
|
Benvenutti R, Gallas-Lopes M, Marcon M, Reschke CR, Herrmann AP, Piato A. Glutamate Nmda Receptor Antagonists With Relevance To Schizophrenia: A Review Of Zebrafish Behavioral Studies. Curr Neuropharmacol 2021; 20:494-509. [PMID: 33588731 PMCID: PMC9608229 DOI: 10.2174/1570159x19666210215121428] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 11/22/2022] Open
Abstract
Schizophrenia pathophysiology is associated with hypofunction of glutamate NMDA receptors (NMDAR) in GABAergic interneurons and dopaminergic hyperactivation in subcortical brain areas. The administration of NMDAR antagonists is used as an animal model that replicates behavioral phenotypes relevant to the positive, negative, and cognitive symptoms of schizophrenia. Such models overwhelmingly rely on rodents, which may lead to species-specific biases and poor translatability. Zebrafish, however, is increasingly used as a model organism to study evolutionarily conserved aspects of behavior. We thus aimed to review and integrate the major findings reported in the zebrafish literature regarding the behavioral effects of NMDAR antagonists with relevance to schizophrenia. We identified 44 research articles that met our inclusion criteria from 590 studies retrieved from MEDLINE (PubMed) and Web of Science databases. Dizocilpine (MK-801) and ketamine were employed in 29 and 10 studies, respectively. The use of other NMDAR antagonists, such as phencyclidine (PCP), APV, memantine, and tiletamine, was described in 6 studies. Frequently reported findings are the social interaction and memory deficits induced by MK-801 and circling behavior induced by ketamine. However, mixed results were described for several locomotor and exploratory parameters in the novel tank and open tank tests. The present review integrates the most relevant results while discussing variation in experimental design and methodological procedures. We conclude that zebrafish is a suitable model organism to study drug-induced behavioral phenotypes relevant to schizophrenia. However, more studies are necessary to further characterize the major differences in behavior as compared to mammals.
Collapse
Affiliation(s)
- Radharani Benvenutti
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS. Brazil
| | - Matheus Gallas-Lopes
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS. Brazil
| | - Matheus Marcon
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS. Brazil
| | - Cristina R Reschke
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin. Ireland
| | - Ana Paula Herrmann
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS. Brazil
| | - Angelo Piato
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS. Brazil
| |
Collapse
|
28
|
Savalia NK, Shao LX, Kwan AC. A Dendrite-Focused Framework for Understanding the Actions of Ketamine and Psychedelics. Trends Neurosci 2020; 44:260-275. [PMID: 33358035 DOI: 10.1016/j.tins.2020.11.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/07/2020] [Accepted: 11/24/2020] [Indexed: 02/09/2023]
Abstract
Pilot studies have hinted that serotonergic psychedelics such as psilocybin may relieve depression, and could possibly do so by promoting neural plasticity. Intriguingly, another psychotomimetic compound, ketamine, is a fast-acting antidepressant and induces synapse formation. The similarities in behavioral and neural effects have been puzzling because the compounds target distinct molecular receptors in the brain. In this opinion article, we develop a conceptual framework that suggests the actions of ketamine and serotonergic psychedelics may converge at the dendrites, to both enhance and suppress membrane excitability. We speculate that mismatches in the opposing actions on dendritic excitability may relate to these compounds' cell-type and region selectivity, their moderate range of effects and toxicity, and their plasticity-promoting capacities.
Collapse
Affiliation(s)
- Neil K Savalia
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Ling-Xiao Shao
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Alex C Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
29
|
Molecular mechanisms of action determine inhibition of paroxysmal depolarizing shifts by NMDA receptor antagonists in rat cortical neurons. Neuropharmacology 2020; 184:108443. [PMID: 33345828 DOI: 10.1016/j.neuropharm.2020.108443] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 01/28/2023]
Abstract
N-methyl-d-aspartate glutamate receptors (NMDARs) are involved in numerous central nervous system (CNS) processes, including epileptiform activity. We used a picrotoxin-induced epileptiform activity model to compare the action of different types of NMDAR antagonists in rat brain slices. Paroxysmal depolarizing shifts (PDS) were evoked by external stimulation in the medial prefrontal cortex (mPFC) slices and recorded in pyramidal cells (PC) and in fast-spiking interneurons (FSI). The NMDAR antagonists APV and memantine reduced the duration of PDS. However, the competitive antagonist APV caused similar effects on the PC and FSI, while the open-channel blocker memantine had a much stronger effect on the PDS in the FSI than in the PC. This difference cannot be explained by a corresponding difference in NMDAR sensitivity to memantine because the drug inhibited the excitatory postsynaptic current (EPSC) similarly in both cell types. Importantly, the PDS were significantly longer in the FSI than in the PC. The degree of PDS inhibition by memantine correlated with individual PDS durations in each cell type. Computer modeling of a synaptic network in the mPFC suggests that the different effects of memantine on the PDS in the PC and FSI can be explained by use dependence of its action. An open-channel blocking mechanism and competition with Mg2+ ions for the binding site result in pronounced inhibition of the long PDS, whereas the short PDS are weakly sensitive. Our results show that peculiarities of kinetics and the mechanism of action largely determine the effects of NMDAR antagonists on physiological and/or pathological processes.
Collapse
|
30
|
Alshammari TK. The Ketamine Antidepressant Story: New Insights. Molecules 2020; 25:molecules25235777. [PMID: 33297563 PMCID: PMC7730956 DOI: 10.3390/molecules25235777] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022] Open
Abstract
Ketamine is a versatile agent primarily utilized as a dissociative anesthetic, which acts by blocking the excitatory receptor N-methyl-d-aspartate receptor (NMDA). It functions to inhibit the current of both Na+ and K+ voltage-gated channels, thus preventing serotonin and dopamine reuptake. Studies have indicated that administering a single subanesthetic dose of ketamine relieves depression rapidly and that the effect is sustained. For decades antidepressant agents were based on the monoamine theory. Although ketamine may not be the golden antidepressant, it has opened new avenues toward mechanisms involved in the pathology of treatment-resistant depression and achieving rapid antidepressant effects. Thus, preclinical studies focusing on deciphering the molecular mechanisms involved in the antidepressant action of ketamine will assist in the development of a new antidepressant. This review was conducted to elucidate the emerging pathways that can explain the complex dose-dependent mechanisms achieved by administering ketamine to treat major depressive disorders. Special attention was paid to reviewing the literature on hydroxynorketamines, which are ketamine metabolites that have recently attracted attention in the context of depression.
Collapse
Affiliation(s)
- Tahani K Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2475, Riyadh 11451, Saudi Arabia
| |
Collapse
|
31
|
Phillips MB, Nigam A, Johnson JW. Interplay between Gating and Block of Ligand-Gated Ion Channels. Brain Sci 2020; 10:brainsci10120928. [PMID: 33271923 PMCID: PMC7760600 DOI: 10.3390/brainsci10120928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/21/2020] [Accepted: 11/26/2020] [Indexed: 02/03/2023] Open
Abstract
Drugs that inhibit ion channel function by binding in the channel and preventing current flow, known as channel blockers, can be used as powerful tools for analysis of channel properties. Channel blockers are used to probe both the sophisticated structure and basic biophysical properties of ion channels. Gating, the mechanism that controls the opening and closing of ion channels, can be profoundly influenced by channel blocking drugs. Channel block and gating are reciprocally connected; gating controls access of channel blockers to their binding sites, and channel-blocking drugs can have profound and diverse effects on the rates of gating transitions and on the stability of channel open and closed states. This review synthesizes knowledge of the inherent intertwining of block and gating of excitatory ligand-gated ion channels, with a focus on the utility of channel blockers as analytic probes of ionotropic glutamate receptor channel function.
Collapse
Affiliation(s)
- Matthew B. Phillips
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (M.B.P.); (A.N.)
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Aparna Nigam
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (M.B.P.); (A.N.)
| | - Jon W. Johnson
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (M.B.P.); (A.N.)
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Correspondence: ; Tel.: +1-(412)-624-4295
| |
Collapse
|
32
|
Ghatak S, Talantova M, McKercher SR, Lipton SA. Novel Therapeutic Approach for Excitatory/Inhibitory Imbalance in Neurodevelopmental and Neurodegenerative Diseases. Annu Rev Pharmacol Toxicol 2020; 61:701-721. [PMID: 32997602 DOI: 10.1146/annurev-pharmtox-032320-015420] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Excitatory/inhibitory (E/I) balance, defined as the balance between excitation and inhibition of synaptic activity in a neuronal network, accounts in part for the normal functioning of the brain, controlling, for example, normal spike rate. In many pathological conditions, this fine balance is perturbed, leading to excessive or diminished excitation relative to inhibition, termed E/I imbalance, reflected in network dysfunction. E/I imbalance has emerged as a contributor to neurological disorders that occur particularly at the extremes of life, including autism spectrum disorder and Alzheimer's disease, pointing to the vulnerability of neuronal networks at these critical life stages. Hence, it is important to develop approaches to rebalance neural networks. In this review, we describe emerging therapies that can normalize the E/I ratio or the underlying abnormality that contributes to the imbalance in electrical activity, thus improving neurological function in these maladies.
Collapse
Affiliation(s)
- Swagata Ghatak
- Department of Molecular Medicine and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, California 92037, USA;
| | - Maria Talantova
- Department of Molecular Medicine and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, California 92037, USA;
| | - Scott R McKercher
- Department of Molecular Medicine and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, California 92037, USA;
| | - Stuart A Lipton
- Department of Molecular Medicine and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, California 92037, USA; .,Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
33
|
Zhuo C, Ji F, Tian H, Wang L, Jia F, Jiang D, Chen C, Zhou C, Lin X, Zhu J. Transient effects of multi-infusion ketamine augmentation on treatment-resistant depressive symptoms in patients with treatment-resistant bipolar depression - An open-label three-week pilot study. Brain Behav 2020; 10:e01674. [PMID: 32621379 PMCID: PMC7428494 DOI: 10.1002/brb3.1674] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/29/2020] [Accepted: 05/08/2020] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION While the psychiatric benefits of ketamine have been verified through clinical trials, there is limited information about ketamine augmentation in patients with treatment-resistant bipolar depression (TRBPD). Hence, in the present study, we investigate the therapeutic efficacy and functional brain alterations associated with multi-infusion ketamine augmentation in patients with TRBPD. METHODS The present three-week study included 38 patients with TRBPD, all of whom received a series of nine ketamine injections over the study period. The Hamilton Depression Rating Scale (HAMD) was used to assess the effects of multi-infusion ketamine combined with mood stabilizers. Brain function was evaluated by global functional connectivity density (gFCD). RESULTS Adjunctive treatment with multiple infusions of ketamine, when combined with a mood stabilizer, could effectively alleviate depressive symptoms for one week, yet the symptoms began to relapse during the second week. Functional brain alterations were detected via gFCD. Specifically, gFCD reductions were mainly found in the bilateral insula, right caudate nucleus, and bilateral inferior frontal gyrus, while increased gFCD was mainly located in the bilateral postcentral gyrus, subgenual anterior cingulate cortex, bilateral thalamus, and cerebellum. Although gFCD alterations were sustained for up to three weeks after the first ketamine infusion, the antidepressant effects of ketamine augmentation sharply declined from the end of the second week of treatment. CONCLUSIONS Multi-infusion ketamine augmentation can rapidly alleviate depressive symptoms in patients with TRBPD. The clinical effects were primarily visible in the first week after treatment and partially sustained for two weeks; however, the therapeutic effects and related functional brain alterations sharply decreased from the end of the second week. Based on these findings, we demonstrated that the clinical efficacy and functional brain alterations induced by ketamine augmentation are transient.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Department of Psychiatry, School of Mental Health, Jining Medical University, Jining, China.,Department of Psychiatry and Imaging-Genetics and Co-morbidity (PNGC-Lab), Tianjin Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China.,Department of Psychiatry Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Feng Ji
- Department of Psychiatry, School of Mental Health, Jining Medical University, Jining, China
| | - Hongjun Tian
- Department of Psychiatry and Imaging-Genetics and Co-morbidity (PNGC-Lab), Tianjin Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Lina Wang
- Department of Psychiatry and Imaging-Genetics and Co-morbidity (PNGC-Lab), Tianjin Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Feng Jia
- Department of Psychiatry and Imaging-Genetics and Co-morbidity (PNGC-Lab), Tianjin Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Deguo Jiang
- Department of Psychiatry Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Ce Chen
- Department of Psychiatry Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Chunhua Zhou
- Department of Pharmacy, First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaodong Lin
- Department of Psychiatry Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Jingjing Zhu
- Department of Psychiatry Wenzhou Seventh People's Hospital, Wenzhou, China
| |
Collapse
|
34
|
Molina JL, Voytek B, Thomas ML, Joshi YB, Bhakta SG, Talledo JA, Swerdlow NR, Light GA. Memantine Effects on Electroencephalographic Measures of Putative Excitatory/Inhibitory Balance in Schizophrenia. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 5:562-568. [PMID: 32340927 PMCID: PMC7286803 DOI: 10.1016/j.bpsc.2020.02.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Abnormalities in cortical excitation and inhibition (E/I) balance are thought to underlie sensory and information processing deficits in schizophrenia. Deficits in early auditory information processing mediate both neurocognitive and functional impairment and appear to be normalized by acute pharmacologic challenge with the NMDA antagonist memantine (MEM). METHODS Thirty-six subjects with a diagnosis of schizophrenia and 31 healthy control subjects underwent electroencephalographic recordings. Subjects ingested either placebo or MEM (10 or 20 mg) in a double-blind, within-subject, crossover, randomized design. The aperiodic, 1/f-like scaling property of the neural power spectra, which is thought to index relative E/I balance, was estimated using a robust linear regression algorithm. RESULTS Patients with schizophrenia had greater aperiodic components compared with healthy control subjects (p < .01, d = 0.64), which was normalized after 20 mg MEM. Analysis revealed a significant dose × diagnosis interaction (p < .0001, d = 0.82). Furthermore, the MEM effect (change in aperiodic component in MEM vs. placebo conditions) was associated with baseline attention and vigilance (r = .54, p < .05) and MEM-induced enhancements in gamma power (r = -.60, p < .01). CONCLUSIONS Findings confirmed E/I balance abnormalities in schizophrenia that were normalized with acute MEM administration and suggest that neurocognitive profiles may predict treatment response based on E/I sensitivity. These data provide proof-of-concept evidence for the utility of E/I balance indices as metrics of acute pharmacologic sensitivity for central nervous system therapeutics.
Collapse
Affiliation(s)
- Juan L Molina
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Bradley Voytek
- Department of Cognitive Sciences, Halicioğlu Data Science Institute, and Neurosciences Graduate Program, University of California, San Diego, La Jolla, California
| | - Michael L Thomas
- Department of Psychiatry, University of California, San Diego, La Jolla, California; Department of Psychology, Colorado State University, Fort Collins, Colorado
| | - Yash B Joshi
- Department of Psychiatry, University of California, San Diego, La Jolla, California; VA Desert Pacific Mental Illness Research, Education and Clinical Center, VA San Diego Healthcare System, San Diego, California
| | - Savita G Bhakta
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Jo A Talledo
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Neal R Swerdlow
- Department of Psychiatry, University of California, San Diego, La Jolla, California.
| | - Gregory A Light
- Department of Psychiatry, University of California, San Diego, La Jolla, California; VA Desert Pacific Mental Illness Research, Education and Clinical Center, VA San Diego Healthcare System, San Diego, California
| |
Collapse
|
35
|
Zhuo C, Lin X, Tian H, Liu S, Bian H, Chen C. Adjunct ketamine treatment of depression in treatment-resistant schizophrenia patients is unsatisfactory in pilot and secondary follow-up studies. Brain Behav 2020; 10:e01600. [PMID: 32174025 PMCID: PMC7218248 DOI: 10.1002/brb3.1600] [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: 09/18/2019] [Revised: 02/07/2020] [Accepted: 02/25/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To investigate the effects of adjunct ketamine treatment on chronic treatment-resistant schizophrenia patients with treatment-resistant depressive symptoms (CTRS-TRD patients), including alterations in brain function. METHODS Intravenous ketamine (0.5 mg/kg body weight) was administered to CTRS-TRD patients over a 1-hr period on days 1, 4, 7, 10, 13, 16, 19, 22, and 25 of our initial pilot study. This treatment method was subsequently repeated 58 days after the start of the pilot study for a secondary follow-up study. Calgary Depression Scale for Schizophrenia (CDSS), Positive and Negative Syndrome Scale (PANSS), and regional homogeneity (ReHo) results were used to assess treatment effects and alterations in brain function throughout the entire duration of our studies. RESULTS Between day 7 and day 14 of the first treatment, CDSS scores were reduced by 63.8% and PANSS scores were reduced by 30.04%. In addition, ReHo values increased in the frontal, temporal, and parietal lobes. However, by day 21, depressive symptoms relapsed. During the second treatment period, CDSS and PANSS scores exhibited no significant differences compared to baseline between day 58 and day 86. On day 65, ReHo values were higher in the temporal, frontal, and parietal lobes. However, on day 79, the increase in ReHo values completely disappeared. CONCLUSIONS Depressive symptoms in CTRS-TRD patients were alleviated with adjunct ketamine treatment for only 1 week during the first treatment period. Moreover, after 1 month, the antidepressant effects of ketamine on CTRS-TRD patients completely disappeared. Correspondingly, ReHo alterations induced by ketamine in the CTRS-TRD patients were not maintained for more than 3 weeks. These pilot findings indicate that adjunct ketamine treatment is not satisfactory for CTRS-TRD patients.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Department of Psychiatry, School of Mental Health, Jining Medical University, Jining, China.,Department of Psychiatric-Neuroimaging-Genetics Laboratory (PNG_Lab), Wenzhou Seventh People's Hospital, Wenzhou, China.,PNGC-Lab, Tianjin Mental Health Centre, Tianjin Anding Hospital, Tianjin, China
| | - Xiaodong Lin
- Department of Psychiatric-Neuroimaging-Genetics Laboratory (PNG_Lab), Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Hongjun Tian
- PNGC-Lab, Tianjin Mental Health Centre, Tianjin Anding Hospital, Tianjin, China
| | - Sha Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Tainyuan, China
| | - Haiman Bian
- Department of Radiology, The Fourth Centre Hospital of Tianjin, Tianjin Medical University Affiliated Fourth Centre Hospital, Tianijn, China
| | - Ce Chen
- Department of Psychiatric-Neuroimaging-Genetics Laboratory (PNG_Lab), Wenzhou Seventh People's Hospital, Wenzhou, China
| |
Collapse
|
36
|
Rantamäki T, Kohtala S. Encoding, Consolidation, and Renormalization in Depression: Synaptic Homeostasis, Plasticity, and Sleep Integrate Rapid Antidepressant Effects. Pharmacol Rev 2020; 72:439-465. [DOI: 10.1124/pr.119.018697] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
37
|
Iacobucci GJ, Popescu GK. Ca 2+-Dependent Inactivation of GluN2A and GluN2B NMDA Receptors Occurs by a Common Kinetic Mechanism. Biophys J 2020; 118:798-812. [PMID: 31629478 PMCID: PMC7036730 DOI: 10.1016/j.bpj.2019.07.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/14/2019] [Accepted: 07/10/2019] [Indexed: 12/12/2022] Open
Abstract
N-Methyl-d-aspartate (NMDA) receptors are Ca2+-permeable channels gated by glutamate and glycine that are essential for central excitatory transmission. Ca2+-dependent inactivation (CDI) is a regulatory feedback mechanism that reduces GluN2A-type NMDA receptor responses in an activity-dependent manner. Although CDI is mediated by calmodulin binding to the constitutive GluN1 subunit, prior studies suggest that GluN2B-type receptors are insensitive to CDI. We examined the mechanism of CDI subtype dependence using electrophysiological recordings of recombinant NMDA receptors expressed in HEK-293 cells. In physiological external Ca2+, we observed robust CDI of whole-cell GluN2A currents (0.42 ± 0.05) but no CDI in GluN2B currents (0.08 ± 0.07). In contrast, when Ca2+ was supplied intracellularly, robust CDI occurred for both GluN2A and GluN2B currents (0.75 ± 0.03 and 0.67 ± 0.02, respectively). To examine how the source of Ca2+ affects CDI, we recorded one-channel Na+ currents to quantify the receptor gating mechanism while simultaneously monitoring ionomycin-induced intracellular Ca2+ elevations with fluorometry. We found that CDI of both GluN2A and GluN2B receptors reflects receptor accumulation in long-lived closed (desensitized) states, suggesting that the observed subtype-dependent differences in macroscopic CDI reflect intrinsic differences in equilibrium open probabilities (Po). We tested this hypothesis by measuring substantial macroscopic CDI, in physiologic conditions, for high Po GluN2B receptors (GluN1A652Y/GluN2B). Together, these results show that Ca2+ flux produces activity-dependent inactivation for both GluN2A and GluN2B receptors and that the extent of CDI varies with channel Po. These results are consistent with CDI as an autoinhibitory feedback mechanism against excessive Ca2+ load during high Po activation.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York.
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York
| |
Collapse
|
38
|
Czerniczyniec A, Karadayian AG, Bustamante J, Lores-Arnaiz S. Ketamine treatment affects hippocampal but not cortical mitochondrial function in prepubertal rats. Int J Dev Neurosci 2020; 80:175-187. [PMID: 32053738 DOI: 10.1002/jdn.10015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/20/2020] [Accepted: 02/07/2020] [Indexed: 12/19/2022] Open
Abstract
Previous reports have shown that ketamine triggered apoptosis in immature developing brain involving mitochondrial-mediated pathways. However, no data for ketamine effects on hippocampal and cortical mitochondrial function are available in prepubertal rats. Twenty-one-day-old Sprague-Dawley rats received ketamine (40 mg/kg i.p.) for 3 days and were killed 24 hr after the last injection. Hippocampal mitochondria from ketamine-treated rats showed decreased malate-glutamate state 4 and 3 respiratory rates and an inhibition in complex I and IV activities. Hippocampal mitochondrial membrane depolarization and mitochondrial permeability transition induction were observed. This was not reflected in an increment of H2 O2 production probably due to increased Mn-SOD and catalase activities, 24 hr after treatment. Interestingly, increased H2 O2 production rates and cardiolipin oxidation were found in hippocampal mitochondria shortly after ketamine treatment (45 min). Unlike the hippocampus, ketamine did not affect mitochondrial parameters in the brain cortex, being the area less vulnerable to suffer ketamine-induced oxidative damage. Results provide evidences that exposure of prepubertal rats to ketamine leads to an induction of mitochondrial ROS generation at early stages of treatment that was normalized by the triggering of antioxidant systems. Although hippocampal mitochondria from prepubertal rats were capable of responding to the oxidative stress, they remain partially dysfunctional.
Collapse
Affiliation(s)
- Analía Czerniczyniec
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Bioquímica y Medicina Molecular (IBIMOL), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía G Karadayian
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Bioquímica y Medicina Molecular (IBIMOL), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juanita Bustamante
- Centro de Altos Estudios en Ciencias de la Salud, Universidad Abierta Interamericana, Buenos Aires, Argentina
| | - Silvia Lores-Arnaiz
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Bioquímica y Medicina Molecular (IBIMOL), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
39
|
Galzitskaya OV. Oligomers Are Promising Targets for Drug Development in the Treatment of Proteinopathies. Front Mol Neurosci 2020; 12:319. [PMID: 32076398 PMCID: PMC7006448 DOI: 10.3389/fnmol.2019.00319] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 12/16/2019] [Indexed: 12/31/2022] Open
Abstract
Currently, there is no effective treatment of proteinopathies, as well as their diagnosis in the early stages of the disease until the first clinical symptoms appear. The proposed model of fibrillation of the Aβ peptide and its fragments not only describes molecular rearrangements, but also offers models of processes that occur during the formation of amyloid aggregates. Since this model is also characteristic of other proteins and peptides, a new potential target for drug development in the treatment of Alzheimer’s disease (AD) and other proteinopathies is proposed on the basis of this model. In our opinion, it is oligomers that are promising targets for innovative developments in the treatment of these diseases.
Collapse
Affiliation(s)
- Oxana V Galzitskaya
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, Pushchino, Russia.,Laboratory of the Structure and Function of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
40
|
Tanaka M, Bohár Z, Vécsei L. Are Kynurenines Accomplices or Principal Villains in Dementia? Maintenance of Kynurenine Metabolism. Molecules 2020; 25:molecules25030564. [PMID: 32012948 PMCID: PMC7036975 DOI: 10.3390/molecules25030564] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/24/2020] [Accepted: 01/25/2020] [Indexed: 12/16/2022] Open
Abstract
Worldwide, 50 million people suffer from dementia, a group of symptoms affecting cognitive and social functions, progressing severely enough to interfere with daily life. Alzheimer’s disease (AD) accounts for most of the dementia cases. Pathological and clinical findings have led to proposing several hypotheses of AD pathogenesis, finding a presence of positive feedback loops and additionally observing the disturbance of a branch of tryptophan metabolism, the kynurenine (KYN) pathway. Either causative or resultant of dementia, elevated levels of neurotoxic KYN metabolites are observed, potentially upregulating multiple feedback loops of AD pathogenesis. Memantine is an N-methyl-D-aspartate glutamatergic receptor (NMDAR) antagonist, which belongs to one of only two classes of medications approved for clinical use, but other NMDAR modulators have been explored so far in vain. An endogenous KYN pathway metabolite, kynurenic acid (KYNA), likewise inhibits the excitotoxic NMDAR. Besides its anti-excitotoxicity, KYNA is a multitarget compound that triggers anti-inflammatory and antioxidant activities. Modifying the KYNA level is a potential multitarget strategy to normalize the disturbed KYN pathway and thus to alleviate juxtaposing AD pathogeneses. In this review, the maintenance of KYN metabolism by modifying the level of KYNA is proposed and discussed in search for a novel lead compound against the progression of dementia.
Collapse
Affiliation(s)
- Masaru Tanaka
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Zsuzsanna Bohár
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - László Vécsei
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-351
| |
Collapse
|
41
|
Nasir M, Trujillo D, Levine J, Dwyer JB, Rupp ZW, Bloch MH. Glutamate Systems in DSM-5 Anxiety Disorders: Their Role and a Review of Glutamate and GABA Psychopharmacology. Front Psychiatry 2020; 11:548505. [PMID: 33329087 PMCID: PMC7710541 DOI: 10.3389/fpsyt.2020.548505] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Serotonin reuptake inhibitors and benzodiazepines are evidence-based pharmacological treatments for Anxiety Disorders targeting serotonin and GABAergic systems, respectively. Although clearly effective, these medications fail to improve anxiety symptoms in a significant proportion of patients. New insights into the glutamate system have directed attention toward drugs that modulate glutamate as potential alternative treatments for anxiety disorders. Here we summarize the current understanding of the potential role of glutamate neurotransmission in anxiety disorders and highlight specific glutamate receptors that are potential targets for novel anxiety disorder treatments. We also review clinical trials of medications targeting the glutamate system in DSM-5 anxiety disorders. Understanding the role of the glutamate system in the pathophysiology of anxiety disorder may aid in developing novel pharmacological agents that are effective in treating anxiety disorders.
Collapse
Affiliation(s)
- Madeeha Nasir
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States
| | - Daniel Trujillo
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States
| | - Jessica Levine
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States
| | - Jennifer B Dwyer
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States.,Yale Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
| | - Zachary W Rupp
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States.,Frank H. Netter School of Medicine, Quinnipiac University, North Haven, CT, United States
| | - Michael H Bloch
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States.,Yale Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
42
|
Dual action of amitriptyline on NMDA receptors: enhancement of Ca-dependent desensitization and trapping channel block. Sci Rep 2019; 9:19454. [PMID: 31857688 PMCID: PMC6923474 DOI: 10.1038/s41598-019-56072-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 12/04/2019] [Indexed: 12/18/2022] Open
Abstract
Although the tricyclic antidepressant amitriptyline (ATL) is widely used in the clinic, the mechanism underlying its high therapeutic efficacy against neuropathic pain remains unclear. NMDA receptors (NMDARs) represent a target for ATL and are involved in sensitization of neuropathic pain. Here we describe two actions of ATL on NMDARs: 1) enhancement of Ca2+-dependent desensitization and 2) trapping channel block. Inhibition of NMDARs by ATL was found to be dependent upon external Ca2+ concentration ([Ca2+]) in a voltage-independent manner, with an IC50 of 0.72 μM in 4 mM [Ca2+]. The ATL IC50 value increased exponentially with decreasing [Ca2+], with an e-fold change observed per 0.69 mM decrease in [Ca2+]. Loading neurons with BAPTA abolished Ca2+-dependent inhibition, suggesting that Ca2+ affects NMDARs from the cytosol. Since there is one known Ca2+-dependent process in gating of NMDARs, we conclude that ATL most likely promotes Ca2+-dependent desensitization. We also found ATL to be a trapping open-channel blocker of NMDARs with an IC50 of 220 µM at 0 mV. An e-fold change in ATL IC50 was observed to occur with a voltage shift of 50 mV in 0.25 mM [Ca2+]. Thus, we disclose here a robust dependence of ATL potency on extracellular [Ca2+], and demonstrate that ATL bound in the NMDAR pore can be trapped by closure of the channel.
Collapse
|
43
|
Bustamante J, Acosta L, Karadayian AG, Lores-Arnaiz S. Ketamine induced cell death can be mediated by voltage dependent calcium channels in PC12 cells. Exp Mol Pathol 2019; 111:104318. [DOI: 10.1016/j.yexmp.2019.104318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 09/27/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023]
|
44
|
Iacobucci GJ, Popescu GK. Spatial Coupling Tunes NMDA Receptor Responses via Ca 2+ Diffusion. J Neurosci 2019; 39:8831-8844. [PMID: 31519826 PMCID: PMC6832682 DOI: 10.1523/jneurosci.0901-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 08/11/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
In the CNS, NMDA receptors generate large and highly regulated Ca2+ signals, which are critical for synaptic development and plasticity. They are highly clustered at postsynaptic sites and along dendritic arbors, but whether this spatial arrangement affects their output is unknown. Synaptic NMDA receptor currents are subject to Ca2+-dependent inactivation (CDI), a type of activity-dependent inhibition that requires intracellular Ca2+ and calmodulin (CaM). We asked whether Ca2+ influx through a single NMDA receptor influences the activity of nearby NMDA receptors, as a possible coupling mechanism. Using cell-attached unitary current recordings from GluN1-2a/GluN2A receptors expressed in human HEK293 cells and from NMDA receptors native to hippocampal neurons from male and female rats, we recorded unitary currents from multichannel patches and used a coupled Markov model to determine the extent of signal coupling (κ). In the absence of extracellular Ca2+, we observed no cooperativity (κ < 0.1), whereas in 1.8 mm external Ca2+, both recombinant and native channels showed substantial negative cooperativity (κ = 0.27). Intracellular Ca2+ chelation or overexpression of a Ca2+-insensitive CaM mutant, reduced coupling, which is consistent with CDI representing the coupling mechanism. In contrast, cooperativity increased substantially (κ = 0.68) when overexpressing the postsynaptic scaffolding protein PSD-95, which increased receptor clustering. Together, these new results demonstrate that NMDA receptor currents are negatively coupled through CDI, and the degree of coupling can be tuned by the distance between receptors. Therefore, channel clustering can influence the activity-dependent reduction in NMDA receptor currents.SIGNIFICANCE STATEMENT At central synapses, NMDA receptors are a major class of excitatory glutamate-gated channels and a source of activity-dependent Ca2+ influx. In turn, fluxed Ca2+ ions bind to calmodulin-primed receptors and reduce further entry, through an autoinhibitory mechanism known as Ca2+ -dependent inactivation (CDI). Here, we show that the diffusion of fluxed Ca2+ between active channels situated within submicroscopic distances amplified receptor inactivation. Thus, calmodulin-mediated gating modulation, an evolutionarily conserved regulatory mechanism, endows synapses with sensitivity to both the temporal sequence and spatial distribution of Ca2+ signals. Perturbations in this mechanism, which coordinates the activity of NMDA receptors within a cluster, may cause signaling alterations that contribute to neuropsychiatric conditions.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14206
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14206
| |
Collapse
|
45
|
Demartini B, Gambini O, Uggetti C, Cariati M, Cadioli M, Goeta D, Marceglia S, Ferrucci R, Priori A. Limbic neurochemical changes in patients with functional motor symptoms. Neurology 2019; 93:e52-e58. [DOI: 10.1212/wnl.0000000000007717] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 02/11/2019] [Indexed: 01/04/2023] Open
Abstract
ObjectiveTo assess by magnetic resonance spectroscopy (MRS) the N-acetylaspartate, myo-inositol, choline, sum of glutamate and glutamine (Glx), and creatine (Cr) content in the anterior cingulate cortex (ACC)/medial prefrontal cortex (mPFC) and in the occipital cortex (OCC) (control region) in patients with functional motor symptoms (FMS) and healthy controls, and to determine whether neurochemical limbic changes as estimated by MRS correlate with FMS-related motor symptom severity, alexithymia, anxiety, depression, and quality of life.MethodsThis case-control study enrolled 10 patients with FMS and 10 healthy controls. Participants underwent MRS and were tested with the Mini-Mental State Examination, Hamilton Depression Rating Scale, Hamilton Anxiety Rating Scale, 20-Item Toronto Alexithymia Scale, and EuroQol 5D.ResultsIn patients with FMS, MRS showed increased Glx/Cr in the ACC/mPFC but normal content in the control OCC. All the other metabolites tested were normal in both regions. The increased Glx/Cr content in the ACC/mPFC correlated with alexithymia, anxiety, and severity of symptoms.ConclusionsThe abnormal limbic Glx increase could have a crucial pathophysiologic role in FMS, possibly by altering limbic-motor interactions, ultimately leading to abnormal movements.
Collapse
|
46
|
Michaëlsson H, Andersson M, Svensson J, Karlsson L, Ehn J, Culley G, Engström A, Bergström N, Savvidi P, Kuhn H, Hanse E, Seth H. The novel antidepressant ketamine enhances dentate gyrus proliferation with no effects on synaptic plasticity or hippocampal function in depressive-like rats. Acta Physiol (Oxf) 2019; 225:e13211. [PMID: 30347138 DOI: 10.1111/apha.13211] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/18/2022]
Abstract
AIM Major depressive disorder is a common and debilitating condition with substantial economic impact. Treatment options, although effective, are aimed at relieving the symptoms with limited disease modification. Ketamine, a commonly used anaesthetic, has received substantial attention as it shows rapid antidepressant effects clinically. We studied the effects of ketamine on hippocampal function and dentate gyrus proliferation in rats showing a depressive-like phenotype. METHODS Adolescent and adult animals were pre-natally exposed to the glucocorticoid analog dexamethasone, and we verified a depressive-like phenotype using behavioural tests, such as the sucrose preference. We subsequently studied the effects of ketamine on hippocampal synaptic transmission, plasticity and dentate gyrus proliferation. In addition, we measured hippocampal glutamate receptor expression. We also tested the ketamine metabolite hydroxynorketamine for NMDA-receptor independent effects. RESULTS Surprisingly, our extensive experimental survey revealed limited effects of ketamine or its metabolite on hippocampal function in control as well as depressive-like animals. We found no effects on synaptic efficacy or induction of long-term potentiation in adolescent and adult animals. Also there was no difference when comparing the dorsal and ventral hippocampus. Importantly, however, ketamine 24 hours prior to experimentation significantly increased the dentate gyrus proliferation, as revealed by Ki-67 immunostaining, in the depressive-like phenotype. CONCLUSION We find limited effects of ketamine on hippocampal glutamatergic transmission. Instead, alterations in dentate gyrus proliferation could explain the antidepressant effects of ketamine.
Collapse
Affiliation(s)
- Henrik Michaëlsson
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Mats Andersson
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Johan Svensson
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Lars Karlsson
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Johan Ehn
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Georgia Culley
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Anders Engström
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Nicklas Bergström
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Parthenia Savvidi
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Hans‐Georg Kuhn
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Eric Hanse
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Henrik Seth
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| |
Collapse
|
47
|
Leiva R, Phillips MB, Turcu AL, Gratacòs-Batlle E, León-García L, Sureda FX, Soto D, Johnson JW, Vázquez S. Pharmacological and Electrophysiological Characterization of Novel NMDA Receptor Antagonists. ACS Chem Neurosci 2018; 9:2722-2730. [PMID: 29767953 DOI: 10.1021/acschemneuro.8b00154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
This work reports the synthesis and pharmacological and electrophysiological evaluation of new N-methyl-d-aspartic acid receptor (NMDAR) channel blocking antagonists featuring polycyclic scaffolds. Changes in the chemical structure modulate the potency and voltage dependence of inhibition. Two of the new antagonists display properties comparable to those of memantine, a clinically approved NMDAR antagonist.
Collapse
Affiliation(s)
- Rosana Leiva
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l’Alimentació i Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Matthew B. Phillips
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Andreea L. Turcu
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l’Alimentació i Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
- Neurophysiology Laboratory, Physiology Unit, Department of Biomedicine, Medical School Universitat de Barcelona, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, and Institut of Neurosciences, 08036 Barcelona, Spain
| | - Esther Gratacòs-Batlle
- Neurophysiology Laboratory, Physiology Unit, Department of Biomedicine, Medical School Universitat de Barcelona, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, and Institut of Neurosciences, 08036 Barcelona, Spain
| | - Lara León-García
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, C./St. Llorenç 21, 43201 Reus, Tarragona, Spain
| | - Francesc X. Sureda
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, C./St. Llorenç 21, 43201 Reus, Tarragona, Spain
| | - David Soto
- Neurophysiology Laboratory, Physiology Unit, Department of Biomedicine, Medical School Universitat de Barcelona, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, and Institut of Neurosciences, 08036 Barcelona, Spain
| | - Jon W. Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l’Alimentació i Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| |
Collapse
|
48
|
Kaniakova M, Kleteckova L, Lichnerova K, Holubova K, Skrenkova K, Korinek M, Krusek J, Smejkalova T, Korabecny J, Vales K, Soukup O, Horak M. 7-Methoxyderivative of tacrine is a ‘foot-in-the-door’ open-channel blocker of GluN1/GluN2 and GluN1/GluN3 NMDA receptors with neuroprotective activity in vivo. Neuropharmacology 2018; 140:217-232. [DOI: 10.1016/j.neuropharm.2018.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/17/2018] [Accepted: 08/07/2018] [Indexed: 10/28/2022]
|
49
|
Hansen KB, Yi F, Perszyk RE, Furukawa H, Wollmuth LP, Gibb AJ, Traynelis SF. Structure, function, and allosteric modulation of NMDA receptors. J Gen Physiol 2018; 150:1081-1105. [PMID: 30037851 PMCID: PMC6080888 DOI: 10.1085/jgp.201812032] [Citation(s) in RCA: 342] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/03/2018] [Indexed: 12/22/2022] Open
Abstract
Hansen et al. review recent structural data that have provided insight into the function and allosteric modulation of NMDA receptors. NMDA-type glutamate receptors are ligand-gated ion channels that mediate a Ca2+-permeable component of excitatory neurotransmission in the central nervous system (CNS). They are expressed throughout the CNS and play key physiological roles in synaptic function, such as synaptic plasticity, learning, and memory. NMDA receptors are also implicated in the pathophysiology of several CNS disorders and more recently have been identified as a locus for disease-associated genomic variation. NMDA receptors exist as a diverse array of subtypes formed by variation in assembly of seven subunits (GluN1, GluN2A-D, and GluN3A-B) into tetrameric receptor complexes. These NMDA receptor subtypes show unique structural features that account for their distinct functional and pharmacological properties allowing precise tuning of their physiological roles. Here, we review the relationship between NMDA receptor structure and function with an emphasis on emerging atomic resolution structures, which begin to explain unique features of this receptor.
Collapse
Affiliation(s)
- Kasper B Hansen
- Department of Biomedical and Pharmaceutical Sciences and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT
| | - Feng Yi
- Department of Biomedical and Pharmaceutical Sciences and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT
| | - Riley E Perszyk
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA
| | - Hiro Furukawa
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Lonnie P Wollmuth
- Departments of Neurobiology & Behavior and Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY
| | - Alasdair J Gibb
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
50
|
Glasgow NG, Wilcox MR, Johnson JW. Effects of Mg 2+ on recovery of NMDA receptors from inhibition by memantine and ketamine reveal properties of a second site. Neuropharmacology 2018; 137:344-358. [PMID: 29793153 PMCID: PMC6050087 DOI: 10.1016/j.neuropharm.2018.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/18/2018] [Accepted: 05/11/2018] [Indexed: 01/19/2023]
Abstract
Memantine and ketamine are NMDA receptor (NMDAR) open channel blockers that are thought to act via similar mechanisms at NMDARs, but exhibit divergent clinical effects. Both drugs act by entering open NMDARs and binding at a site deep within the ion channel (the deep site) at which the endogenous NMDAR channel blocker Mg2+ also binds. Under physiological conditions, Mg2+ increases the IC50s of memantine and ketamine through competition for binding at the deep site. Memantine also can inhibit NMDARs after associating with a second site accessible in the absence of agonist, a process termed second site inhibition (SSI) that is not observed with ketamine. Here we investigated the effects of 1 mM Mg2+ on recovery from inhibition by memantine and ketamine, and on memantine SSI, of the four main diheteromeric NMDAR subtypes. We found that: recovery from memantine inhibition depended strongly on the concentration of memantine used to inhibit the NMDAR response; Mg2+ accelerated recovery from memantine and ketamine inhibition through distinct mechanisms and in an NMDAR subtype-dependent manner; and Mg2+ occupation of the deep site disrupted memantine SSI in a subtype-dependent manner. Our results support the hypothesis that memantine associates with, but does not inhibit at the second site. After associating with the second site, memantine can either slowly dissociate directly to the extracellular solution, or transit to the deep site, resulting in typical channel block. Memantine's relatively slow dissociation from the second site underlies the dependence of NMDAR recovery from inhibition on both memantine concentration and on Mg2+.
Collapse
Affiliation(s)
- Nathan G Glasgow
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Madeleine R Wilcox
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Jon W Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|