1
|
Piniella D, Zafra F. Functional crosstalk of the glycine transporter GlyT1 and NMDA receptors. Neuropharmacology 2023; 232:109514. [PMID: 37003571 DOI: 10.1016/j.neuropharm.2023.109514] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
NMDA-type glutamate receptors (NMDARs) constitute one of the main glutamate (Glu) targets in the central nervous system and are involved in synaptic plasticity, which is the molecular substrate of learning and memory. Hypofunction of NMDARs has been associated with schizophrenia, while overstimulation causes neuronal death in neurodegenerative diseases or in stroke. The function of NMDARs requires coincidental binding of Glu along with other cellular signals such as neuronal depolarization, and the presence of other endogenous ligands that modulate their activity by allosterism. Among these allosteric modulators are zinc, protons and Gly, which is an obligatory co-agonist. These characteristics differentiate NMDARs from other receptors, and their structural bases have begun to be established in recent years. In this review we focus on the crosstalk between Glu and glycine (Gly), whose concentration in the NMDAR microenvironment is maintained by various Gly transporters that remove or release it into the medium in a regulated manner. The GlyT1 transporter is particularly involved in this task, and has become a target of great interest for the treatment of schizophrenia since its inhibition leads to an increase in synaptic Gly levels that enhances the activity of NMDARs. However, the only drug that has completed phase III clinical trials did not yield the expected results. Notwithstanding, there are additional drugs that continue to be investigated, and it is hoped that knowledge gained from the recently published 3D structure of GlyT1 may allow the rational design of more effective new drugs.
Collapse
Affiliation(s)
- Dolores Piniella
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain; IdiPAZ, Institute of Health Carlos III (ISCIII), Spain
| | - Francisco Zafra
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain; IdiPAZ, Institute of Health Carlos III (ISCIII), Spain.
| |
Collapse
|
2
|
Deutsch SI, Burket JA. From Mouse to Man: N-Methyl-d-Aspartic Acid Receptor Activation as a Promising Pharmacotherapeutic Strategy for Autism Spectrum Disorders. Med Clin North Am 2023; 107:101-117. [PMID: 36402493 DOI: 10.1016/j.mcna.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The BALB/c mouse displays hypersensitivity to behavioral effects of MK-801 (dizocilpine), a noncompetitive N-methyl-d-aspartic acid (NMDA) receptor "open-channel" blocker, and shows both no preference for an enclosed stimulus mouse over an inanimate object and reduced social interaction with a freely behaving stimulus mouse. NMDA receptor agonist interventions improved measures of social preference and social interaction of the BALB/c mouse model of autism spectrum disorder (ASD). A "proof of principle/proof of concept" translational 10-week clinical trial with 8-week of active medication administration was conducted comparing 20 DSM-IV-TR-diagnosed older adolescent/young adult patients with ASD randomized to once-weekly pulsed administration (50 mg/d) versus daily administration of d-cycloserine (50 mg/d). The results showed that d-cycloserine, a partial glycine agonist, was well tolerated, the 2 dosing strategies did not differ, and improvement was noted on the "lethargy/social withdrawal" and "stereotypic behavior" subscales of the Aberrant Behavior Checklist. NMDA receptor activation contributes to the regulation of mTOR signaling, a pathologic point of convergence in several monogenic syndromic forms of ASD. Furthermore, both NMDA receptor hypofunction and imbalance between NMDA receptor activation mediated by GluN2B and GluN2A-containing NMDA receptors occur as "downstream" consequences of several genetically unrelated abnormalities associated with ASD. NMDA receptor-subtype selective "positive allosteric modulators (PAMs)" are particularly appealing medication candidates for future translational trials.
Collapse
Affiliation(s)
- Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA 23507, USA
| | - Jessica A Burket
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA.
| |
Collapse
|
3
|
Deutsch SI, Luyo ZNM, Burket JA. Targeted NMDA Receptor Interventions for Autism: Developmentally Determined Expression of GluN2B and GluN2A-Containing Receptors and Balanced Allosteric Modulatory Approaches. Biomolecules 2022; 12:biom12020181. [PMID: 35204682 PMCID: PMC8961601 DOI: 10.3390/biom12020181] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/27/2022] Open
Abstract
Various ASD risk alleles have been associated with impairment of NMDA receptor activation (i.e., NMDA Receptor Hypofunction) and/or disturbance of the careful balance between activation mediated by GluN2B-subtype and GluN2A-subtype-containing NMDA receptors. Importantly, although these various risk alleles affect NMDA receptor activation through different mechanisms, they share the pathogenic consequences of causing disturbance of highly regulated NMDA receptor activation. Disturbances of NMDA receptor activation due to sequence variants, protein termination variants and copy number variants are often cell-specific and regionally selective. Thus, translational therapeutic NMDA receptor agonist interventions, which may require chronic administration, must have specificity, selectivity and facilitate NMDA receptor activation in a manner that is physiologic (i.e., mimicking that of endogenously released glutamate and glycine/D-serine released in response to salient and relevant socio-cognitive provocations within discrete neural circuits). Importantly, knockout mice with absent expression and mice with haploinsufficient expression of the deleterious genes often serve as good models to test the potential efficacy of promising pharmacotherapeutic strategies. The Review considers diverse examples of “illness” genes, their pathogenic effects on NMDA receptor activation and, when available, results of studies of impaired sociability in mouse models, including “proof of principle/proof of concept” experiments exploring NMDA receptor agonist interventions and the development of promising positive allosteric modulators (PAMs), which serve as support and models for developing an inventory of PAMs and negative allosteric modulators (NAMs) for translational therapeutic intervention. Conceivably, selective PAMs and NAMs either alone or in combination will be administered to patients guided by their genotype in order to potentiate and/or restore disrupted balance between activation mediated by GluN2B-subtype and GluN2A-subtype containing NMDA receptors.
Collapse
Affiliation(s)
- Stephen I. Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA 23507, USA;
| | - Zachary N. M. Luyo
- Program in Neuroscience, Christopher Newport University, Newport News, VA 23606, USA;
| | - Jessica A. Burket
- Program in Neuroscience, Christopher Newport University, Newport News, VA 23606, USA;
- Department of Molecular Biology & Chemistry, Christopher Newport University, Newport News, VA 23606, USA
- Correspondence: ; Tel.: +1-757-594-8743
| |
Collapse
|
4
|
Burket JA, Matar M, Fesshaye A, Pickle JC, Britten RA. Exposure to Low (≤10 cGy) Doses of 4He Particles Leads to Increased Social Withdrawal and Loss of Executive Function Performance. Radiat Res 2021; 196:345-354. [PMID: 34270762 DOI: 10.1667/rade-20-00251.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 07/06/2021] [Indexed: 11/03/2022]
Abstract
Astronauts on the planned mission to Mars will be exposed to galactic cosmic radiation (GCR), with proton and He particles accounting (in approximately equal amounts) for ∼75% of the equivalent dose. Exposure to ≤15 cGy of space radiation ions with Z ≥ 15 particles has been shown to impair various executive functions, including attentional set shifting and creative problem-solving in rats. Executive functions also regulate social interactions and mood. Should space radiation exposure alter these executive functions as it does cognitive flexibility, there is the possibility of altered interactions among crew members and team cooperativity during prolonged space exploration. This study characterized the effects of ≤10 cGy 400 MeV/n of 4He particles on cognitive flexibility and social interaction (within freely interacting dyads) in male Wistar rats. Exposure to ≥1 cGy 4He ions induced deficits in the SD and/or CD stages of the attentional set shifting (ATSET) task, as reported after exposure to Z ≥ 15 space radiation ions. Should similar effects occur in astronauts, these data suggest that they would have a reduced ability to identify key events in a new situation and would be more easily distracted by extraneous variables. The irradiated rats were also screened for performance in a task for unconstrained cognitive flexibility (UCFlex), often referred to as creative problem-solving. There was a marked dose-dependent change in UCFlex performance with ∼30% of rats exposed to 10 cGy being unable to solve the problem, while the remaining rats took longer than the sham-irradiated animals to resolve the problem. Importantly, performance in the ATSET test was not indicative of UCFlex performance. From a risk assessment perspective, these findings suggest that a value based on a single behavioral end point may not fully represent the cognitive deficits induced by space radiation, even within the cognitive flexibility domain. Rats that received 5 cGy 4He ion irradiation had a significantly lower level of interaction toward their sham-irradiated partners in a non-anxiogenic (uncaged) dyad interactions study. This is consistent with the social withdrawal previously observed in space radiation-exposed male mice in a three-chamber test. 4He-irradiated rats exhibited a significantly higher incidence and duration of self-grooming, which is even more concerning, given that their dyad partners were able to physically interact with the irradiated rats (i.e., touching/climbing over them). This study has established that exposure of male rats to "light" ions such as He affects multiple executive functions resulting in deficits in both sociability and cognitive flexibility, and possibly affective behavior (reward valuation). Further studies are needed to determine if these space radiation-induced co-morbidities are concomitantly induced within individual rats.
Collapse
Affiliation(s)
- Jessica A Burket
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, Virginia 23606
- Department of Program in Neuroscience, Christopher Newport University, Newport News, Virginia 23606
| | - Mona Matar
- National Aeronautics and Space Administration, John H. Glenn Research Center, Cleveland, Ohio 44135
| | - Arriyam Fesshaye
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Jerrah C Pickle
- School of Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Leroy T Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Center for Integrative Neuroinflammatory and Inflammatory diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
5
|
Reichard HA, Schiffer HH, Monenschein H, Atienza JM, Corbett G, Skaggs AW, Collia DR, Ray WJ, Serrats J, Bliesath J, Kaushal N, Lam BP, Amador-Arjona A, Rahbaek L, McConn DJ, Mulligan VJ, Brice N, Gaskin PLR, Cilia J, Hitchcock S. Discovery of TAK-041: a Potent and Selective GPR139 Agonist Explored for the Treatment of Negative Symptoms Associated with Schizophrenia. J Med Chem 2021; 64:11527-11542. [PMID: 34260228 DOI: 10.1021/acs.jmedchem.1c00820] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The orphan G-protein-coupled receptor GPR139 is highly expressed in the habenula, a small brain nucleus that has been linked to depression, schizophrenia (SCZ), and substance-use disorder. High-throughput screening and a medicinal chemistry structure-activity relationship strategy identified a novel series of potent and selective benzotriazinone-based GPR139 agonists. Herein, we describe the chemistry optimization that led to the discovery and validation of multiple potent and selective in vivo GPR139 agonist tool compounds, including our clinical candidate TAK-041, also known as NBI-1065846 (compound 56). The pharmacological characterization of these GPR139 agonists in vivo demonstrated GPR139-agonist-dependent modulation of habenula cell activity and revealed consistent in vivo efficacy to rescue social interaction deficits in the BALB/c mouse strain. The clinical GPR139 agonist TAK-041 is being explored as a novel drug to treat negative symptoms in SCZ.
Collapse
Affiliation(s)
- Holly A Reichard
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Hans H Schiffer
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Holger Monenschein
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Josephine M Atienza
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Gerard Corbett
- Takeda Cambridge Ltd., 418 Cambridge Science Park, Cambridge, Cambridgeshire CB4 0PZ, U.K
| | - Alton W Skaggs
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Deanna R Collia
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - William J Ray
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Jordi Serrats
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Joshua Bliesath
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Nidhi Kaushal
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Betty P Lam
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Alejandro Amador-Arjona
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Lisa Rahbaek
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Donavon J McConn
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Victoria J Mulligan
- Takeda Cambridge Ltd., 418 Cambridge Science Park, Cambridge, Cambridgeshire CB4 0PZ, U.K
| | - Nicola Brice
- Takeda Cambridge Ltd., 418 Cambridge Science Park, Cambridge, Cambridgeshire CB4 0PZ, U.K
| | - Philip L R Gaskin
- Takeda Cambridge Ltd., 418 Cambridge Science Park, Cambridge, Cambridgeshire CB4 0PZ, U.K
| | - Jackie Cilia
- Takeda Cambridge Ltd., 418 Cambridge Science Park, Cambridge, Cambridgeshire CB4 0PZ, U.K
| | - Stephen Hitchcock
- Takeda California, Inc., 9625 Towne Centre Drive, San Diego, California 92121, United States
| |
Collapse
|
6
|
Burket JA, Pickle JC, Rusk AM, Haynes BA, Sharp JA, Deutsch SI. Glycine transporter type 1 (GlyT1) inhibition improves conspecific-provoked immobility in BALB/c mice: Analysis of corticosterone response and glucocorticoid gene expression in cortex and hippocampus. Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109869. [PMID: 31962186 DOI: 10.1016/j.pnpbp.2020.109869] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/15/2020] [Indexed: 12/14/2022]
Abstract
Stress reactivity and glucocorticoid signaling alterations are reported in mouse models of autism spectrum disorder (ASD). BALB/c mice display decreased locomotor activity in the presence of stimulus mice and spend less time exploring enclosed stimulus mice; this mouse strain has been validated as an ASD model. VU0410120, a glycine type 1 transporter (GlyT1) inhibitor, improved sociability in BALB/c mice, consistent with data that NMDA Receptor (NMDAR) activation regulates sociability, and the endogenous tone of NMDAR-mediated neurotransmission is altered in this strain. Effects of a prosocial dose of VU0410120 on conspecific-provoked immobility, and relationships between conspecific-provoked immobility and corticosterone response were explored. VU0410120-treated BALB/c mice showed reduced immobility in the presence of conspecifics and increased the conspecific-provoked corticosterone response. However, the intensity of conspecific-provoked immobility in VU0410120-treated BALB/c mice did not differ as a function of corticosterone response. Expression profiles of 88 glucocorticoid signaling associated genes within frontal cortex and hippocampus were examined. BALB/c mice resistant to prosocial effects of VU0410120 had increased mRNA expression of Ddit4, a negative regulator of mTOR signaling. Dysregulated mTOR signaling activity is a convergent finding in several monogenic syndromic forms of ASD. Prosocial effects of VU0410120 in the BALB/c strain may be related to regulatory influences of NMDAR-activation on mTOR signaling activity. Because corticosterone response is a marker of social stress, the current data suggest that the stressfulness of a social encounter alone may not be the sole determinant of increased immobility in BALB/c mice; this strain may also display an element of social disinterest.
Collapse
Affiliation(s)
- Jessica A Burket
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Jerrah C Pickle
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Allison M Rusk
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Bronson A Haynes
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Julia A Sharp
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States; Anne Armistead Robinson Endowed Chair in Psychiatry, Department of Psychiatry and Behavioral Sciences, 825 Fairfax Avenue, Suite 710, Norfolk, VA 23507, United States.
| |
Collapse
|
7
|
Deutsch SI, Burket JA. An Evolving Therapeutic Rationale for Targeting the α 7 Nicotinic Acetylcholine Receptor in Autism Spectrum Disorder. Curr Top Behav Neurosci 2020; 45:167-208. [PMID: 32468495 DOI: 10.1007/7854_2020_136] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Abnormalities of cholinergic nuclei, cholinergic projections, and cholinergic receptors, as well as abnormalities of growth factors involved in the maturation and maintenance of cholinergic neurons, have been described in postmortem brains of persons with autism spectrum disorder (ASD). Further, microdeletions of the 15q13.3 locus that encompasses CHRNA7, the gene coding the α7 nicotinic acetylcholine receptor (α7 nAChR), are associated with a spectrum of neurodevelopmental disorders, including ASD. The heterozygous 15q13.3 microdeletion syndrome suggests that diminished or impaired transduction of the acetylcholine (ACh) signal by the α7 nAChR can be a pathogenic mechanism of ASD. The α7 nAChR has a role in regulating the firing and function of parvalbumin (PV)-expressing GABAergic projections, which synchronize the oscillatory output of assemblies of pyramidal neurons onto which they project. Synchronous oscillatory output is an electrophysiological substrate for higher executive functions, such as working memory, and functional connectivity between discrete anatomic areas of the brain. The α7 nAChR regulates PV expression and works cooperatively with the co-expressed NMDA receptor in subpopulations of GABAergic interneurons in mouse models of ASD. An evolving literature supports therapeutic exploration of selectively targeted cholinergic interventions for the treatment of ASD, especially compounds that target the α7 nAChR subtype. Importantly, development and availability of high-affinity, brain-penetrable, α7 nAChR-selective agonists, partial agonists, allosteric agonists, and positive allosteric modulators (PAMs) should facilitate "proof-of-principle/concept" clinical trials. nAChRs are pentameric allosteric proteins that function as ligand-gated ion channel receptors constructed from five constituent polypeptide subunits, all of which share a common structural motif. Importantly, in addition to α7 nAChR-gated Ca2+ conductance causing membrane depolarization, there are emerging data consistent with possible metabotropic functions of this ionotropic receptor. The ability of α7-selective type II PAMs to "destabilize" the desensitized state and promote ion channel opening may afford them therapeutic advantages over orthosteric agonists. The current chapter reviews historic and recent literature supporting selective therapeutic targeting of the α7 nAChR in persons affected with ASD.
Collapse
Affiliation(s)
- Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, USA.
| | - Jessica A Burket
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA, USA
| |
Collapse
|
8
|
The Role of the N-Methyl-D-Aspartate Receptors in Social Behavior in Rodents. Int J Mol Sci 2019; 20:ijms20225599. [PMID: 31717513 PMCID: PMC6887971 DOI: 10.3390/ijms20225599] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 01/09/2023] Open
Abstract
The appropriate display of social behaviors is essential for the well-being, reproductive success and survival of an individual. Deficits in social behavior are associated with impaired N-methyl-D-aspartate (NMDA) receptor-mediated neurotransmission. In this review, we describe recent studies using genetically modified mice and pharmacological approaches which link the impaired functioning of the NMDA receptors, especially of the receptor subunits GluN1, GluN2A and GluN2B, to abnormal social behavior. This abnormal social behavior is expressed as impaired social interaction and communication, deficits in social memory, deficits in sexual and maternal behavior, as well as abnormal or heightened aggression. We also describe the positive effects of pharmacological stimulation of the NMDA receptors on these social deficits. Indeed, pharmacological stimulation of the glycine-binding site either by direct stimulation or by elevating the synaptic glycine levels represents a promising strategy for the normalization of genetically-induced, pharmacologically-induced or innate deficits in social behavior. We emphasize on the importance of future studies investigating the role of subunit-selective NMDA receptor ligands on different types of social behavior to provide a better understanding of the underlying mechanisms, which might support the development of selective tools for the optimized treatment of disorders associated with social deficits.
Collapse
|
9
|
Metabotropic functions of the NMDA receptor and an evolving rationale for exploring NR2A-selective positive allosteric modulators for the treatment of autism spectrum disorder. Prog Neuropsychopharmacol Biol Psychiatry 2019; 90:142-160. [PMID: 30481555 DOI: 10.1016/j.pnpbp.2018.11.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/19/2018] [Accepted: 11/22/2018] [Indexed: 12/18/2022]
Abstract
NMDA receptors are widely distributed throughout the brain and major therapeutic challenges include targeting specific NMDA receptor subtypes while preserving spatial and temporal specificity during their activation. The NR2A-subunit containing NMDA receptor is implicated in regulating synchronous oscillatory output of cortical pyramidal neurons, which may be disturbed in clinical presentations of autism spectrum disorder (ASD). Because NR2A-selective positive allosteric modulators (PAMs) preserve spatial and temporal selectivity while activating this subpopulation of receptors, they represent a promising strategy to address neocortical circuit abnormalities in ASD. In addition to promoting Ca2+ entry and membrane depolarization, diverse metabotropic effects of NMDA receptor activation on signal transduction pathways occur within the cell, some of which depend on alignment of protein binding partners. For example, NMDA receptor agonist interventions attenuate impaired sociability in transgenic mice with 'loss-of-function' mutations of the Shank family of scaffolding proteins, which highlights the necessity of a carefully orchestrated alignment of protein binding partners in the excitatory synapse. The current Review considers metabotropic functions of the NMDA receptor that could play a role in sociability and the pathogenesis of ASD (e.g., mTOR signaling), in addition to its more familiar ionotropic functions, and provides a rationale for therapeutic exploration of NR2A-selective PAMs.
Collapse
|
10
|
Um SM, Ha S, Lee H, Kim J, Kim K, Shin W, Cho YS, Roh JD, Kang J, Yoo T, Noh YW, Choi Y, Bae YC, Kim E. NGL-2 Deletion Leads to Autistic-like Behaviors Responsive to NMDAR Modulation. Cell Rep 2018; 23:3839-3851. [DOI: 10.1016/j.celrep.2018.05.087] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/13/2018] [Accepted: 05/25/2018] [Indexed: 01/01/2023] Open
|
11
|
Cioffi CL, Liu S, Wolf MA, Guzzo PR, Sadalapure K, Parthasarathy V, Loong DTJ, Maeng JH, Carulli E, Fang X, Karunakaran K, Matta L, Choo SH, Panduga S, Buckle RN, Davis RN, Sakwa SA, Gupta P, Sargent BJ, Moore NA, Luche MM, Carr GJ, Khmelnitsky YL, Ismail J, Chung M, Bai M, Leong WY, Sachdev N, Swaminathan S, Mhyre AJ. Synthesis and Biological Evaluation of N-((1-(4-(Sulfonyl)piperazin-1-yl)cycloalkyl)methyl)benzamide Inhibitors of Glycine Transporter-1. J Med Chem 2016; 59:8473-94. [PMID: 27559615 DOI: 10.1021/acs.jmedchem.6b00914] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We previously disclosed the discovery of rationally designed N-((1-(4-(propylsulfonyl)piperazin-1-yl)cycloalkyl)methyl)benzamide inhibitors of glycine transporter-1 (GlyT-1), represented by analogues 10 and 11. We describe herein further structure-activity relationship exploration of this series via an optimization strategy that primarily focused on the sulfonamide and benzamide appendages of the scaffold. These efforts led to the identification of advanced leads possessing a desirable balance of excellent in vitro GlyT-1 potency and selectivity, favorable ADME and in vitro pharmacological profiles, and suitable pharmacokinetic and safety characteristics. Representative analogue (+)-67 exhibited robust in vivo activity in the cerebral spinal fluid glycine biomarker model in both rodents and nonhuman primates. Furthermore, rodent microdialysis experiments also demonstrated that oral administration of (+)-67 significantly elevated extracellular glycine levels within the medial prefrontal cortex (mPFC).
Collapse
Affiliation(s)
- Christopher L Cioffi
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Shuang Liu
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Mark A Wolf
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Peter R Guzzo
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Kashinath Sadalapure
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Visweswaran Parthasarathy
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - David T J Loong
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Jun-Ho Maeng
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Edmund Carulli
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Xiao Fang
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Kalesh Karunakaran
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Lakshman Matta
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Sok Hui Choo
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Shailijia Panduga
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Ronald N Buckle
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Randall N Davis
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Samuel A Sakwa
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Priya Gupta
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Bruce J Sargent
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Nicholas A Moore
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Michele M Luche
- Bothell Research Center, AMRI , 22215 26th Ave SE, Bothell, Washington 98021-4425, United States
| | - Grant J Carr
- Bothell Research Center, AMRI , 22215 26th Ave SE, Bothell, Washington 98021-4425, United States
| | - Yuri L Khmelnitsky
- Drug Metabolism and Pharmacokinetics, AMRI , East Campus, 17 University Place, Rensselaer, New York 12144, United States
| | - Jiffry Ismail
- Drug Metabolism and Pharmacokinetics, AMRI , East Campus, 17 University Place, Rensselaer, New York 12144, United States
| | - Mark Chung
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Mei Bai
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Wei Yee Leong
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Nidhi Sachdev
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Srividya Swaminathan
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Andrew J Mhyre
- Bothell Research Center, AMRI , 22215 26th Ave SE, Bothell, Washington 98021-4425, United States
| |
Collapse
|
12
|
Green TL, Burket JA, Deutsch SI. Age-dependent effects on social interaction of NMDA GluN2A receptor subtype-selective antagonism. Brain Res Bull 2016; 125:159-67. [PMID: 27378651 DOI: 10.1016/j.brainresbull.2016.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 11/19/2022]
Abstract
NMDA receptor-mediated neurotransmission is implicated in the regulation of normal sociability in mice. The heterotetrameric NMDA receptor is composed of two obligatory GluN1 and either two "modulatory" GluN2A or GluN2B receptor subunits. GluN2A and GluN2B-containing receptors differ in terms of their developmental expression, distribution between synaptic and extrasynaptic locations, and channel kinetic properties, among other differences. Because age-dependent differences in disruptive effects of GluN2A and GluN2B subtype-selective antagonists on sociability and locomotor activity have been reported in rats, the current investigation explored age-dependent effects of PEAQX, a GluN2A subtype-selective antagonist, on sociability, stereotypic behaviors emerging during social interaction, and spatial working memory in 4- and 8-week old male Swiss Webster mice. The data implicate an age-dependent contribution of GluN2A-containing NMDA receptors to the regulation of normal social interaction in mice. Specifically, at a dose of PEAQX devoid of any effect on locomotor activity and mouse rotarod performance, the social interaction of 8-week old mice was disrupted without any effect on the social salience of a stimulus mouse. Moreover, PEAQX attenuated stereotypic behavior emerging during social interaction in 4- and 8-week old mice. However, PEAQX had no effect on spontaneous alternations, a measure of spatial working memory, suggesting that neural circuits mediating sociability and spatial working memory may be discrete and dissociable from each other. Also, the data suggest that the regulation of stereotypic behaviors and sociability may occur independently of each other. Because expression of GluN2A-containing NMDA receptors occurs at a later developmental stage, they may be more involved in mediating the pathogenesis of ASDs in patients with histories of "regression" after a period of normal development than GluN2B receptors.
Collapse
Affiliation(s)
- Torrian L Green
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA, 23507, United States
| | - Jessica A Burket
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA, 23507, United States
| | - Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA, 23507, United States.
| |
Collapse
|
13
|
Burket JA, Young CM, Green TL, Benson AD, Deutsch SI. Characterization of gait and olfactory behaviors in the Balb/c mouse model of autism spectrum disorders. Brain Res Bull 2016; 122:29-34. [PMID: 26917431 DOI: 10.1016/j.brainresbull.2016.02.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/17/2016] [Accepted: 02/19/2016] [Indexed: 10/22/2022]
Abstract
Abnormalities of gait and olfaction have been reported in persons with autism spectrum disorders (ASDs), which could reflect involvement of the cerebellum and nodes related to olfaction (e.g., olfactory bulb and ventral temporal olfactory cortex) in neural circuits subserving social, cognitive, and motor domains of psychopathology in these disorders. We hypothesized that the Balb/c mouse model of ASD would express "abnormalities" of gait and olfaction, relative to the Swiss Webster comparator strain. Contrary to expectation, Balb/c and Swiss Webster mice did not differ in terms of quantitative measurements of gait and mouse rotarod behavior, and Balb/c mice displayed a shorter latency to approach an unscented cotton swab, suggesting that there was no disturbance of its locomotor behavior. However, Balb/c mice showed significant inhibition of locomotor activity in the presence of floral scents, including novel and familiar floral scents, and a socially salient odor (i.e., concentrated mouse urine); the inhibitory effect on the locomotor behavior of the Balb/c mouse was especially pronounced with the salient social odor. Unlike the Swiss Webster strain, mouse urine lacks social salience for the Balb/c mouse strain, a model of ASD, which does not appear to be an artifact of diminished olfactory sensitivity or impaired locomotion.
Collapse
Affiliation(s)
- Jessica A Burket
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | | | - Torrian L Green
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Andrew D Benson
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States.
| |
Collapse
|
14
|
Shen HY, van Vliet EA, Bright KA, Hanthorn M, Lytle NK, Gorter J, Aronica E, Boison D. Glycine transporter 1 is a target for the treatment of epilepsy. Neuropharmacology 2015; 99:554-65. [PMID: 26302655 PMCID: PMC4655139 DOI: 10.1016/j.neuropharm.2015.08.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/27/2015] [Accepted: 08/19/2015] [Indexed: 11/17/2022]
Abstract
Glycine is the major inhibitory neurotransmitter in brainstem and spinal cord, whereas in hippocampus glycine exerts dual modulatory roles on strychnine-sensitive glycine receptors and on the strychnine-insensitive glycineB site of the N-methyl-D-aspartate receptor (NMDAR). In hippocampus, the synaptic availability of glycine is largely under control of glycine transporter 1 (GlyT1). Since epilepsy is a disorder of disrupted network homeostasis affecting the equilibrium of various neurotransmitters and neuromodulators, we hypothesized that changes in hippocampal GlyT1 expression and resulting disruption of glycine homeostasis might be implicated in the pathophysiology of epilepsy. Using two different rodent models of temporal lobe epilepsy (TLE)--the intrahippocampal kainic acid model of TLE in mice, and the rat model of tetanic stimulation-induced TLE--we first demonstrated robust overexpression of GlyT1 in the hippocampal formation, suggesting dysfunctional glycine signaling in epilepsy. Overexpression of GlyT1 in the hippocampal formation was corroborated in human TLE samples by quantitative real time PCR. In support of a role of dysfunctional glycine signaling in the pathophysiology of epilepsy, both the genetic deletion of GlyT1 in hippocampus and the GlyT1 inhibitor LY2365109 increased seizure thresholds in mice. Importantly, chronic seizures in the mouse model of TLE were robustly suppressed by systemic administration of the GlyT1 inhibitor LY2365109. We conclude that GlyT1 overexpression in the epileptic brain constitutes a new target for therapeutic intervention, and that GlyT1 inhibitors constitute a new class of antiictogenic drugs. These findings are of translational value since GlyT1 inhibitors are already in clinical development to treat cognitive symptoms in schizophrenia.
Collapse
Affiliation(s)
- Hai-Ying Shen
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Kerry-Ann Bright
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA
| | - Marissa Hanthorn
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA
| | - Nikki K Lytle
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA
| | - Jan Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands; SEIN - Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands
| | - Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA.
| |
Collapse
|
15
|
Deutsch SI, Burket JA, Benson AD, Urbano MR. NMDA agonists for autism spectrum disorders: progress and possibilities. FUTURE NEUROLOGY 2015. [DOI: 10.2217/fnl.15.28] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Transgenic mice with NMDA-receptor (NMDAR) hypofunction display impaired sociability (i.e., diminished preference for exploring conspecifics), supporting a critical role for the NMDAR in regulation of sociability. The endogenous tone of NMDAR-mediated neurotransmission is altered in the Balb/c mouse model of autism spectrum disorders; thus, the effects of targeting the NMDAR in Balb/c mice on sociability, cognition and stereotypic behavior were explored. Positive effects of this pharmacotherapeutic strategy were observed in Balb/c and other relevant mouse strains displaying impaired sociability. Furthermore, in a preliminary translational clinical trial, D-cycloserine, a partial glycineB site agonist, improved sociability and diminished stereotypies in a sample of adolescents and young adults with autism spectrum disorders. The data encourage pharmacotherapeutic targeting of the NMDAR.
Collapse
Affiliation(s)
- Stephen I Deutsch
- Department of Psychiatry & Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA 23507–1912, USA
| | - Jessica A Burket
- Department of Psychiatry & Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA 23507–1912, USA
| | - Andrew D Benson
- Department of Psychiatry & Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA 23507–1912, USA
| | - Maria R Urbano
- Department of Psychiatry & Behavioral Sciences, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 710, Norfolk, VA 23507–1912, USA
| |
Collapse
|