Synaptic release of glutamate coincident with postsynaptic depolarization allows NMDA receptors to mediate a slow, Ca2+-permeable component of the excitatory synaptic current. The resulting influx of Ca2+ can trigger changes in synaptic strength that have been proposed as a cellular correlate of learning and memory. Reduction of NMDA receptor function blocks many forms of synaptic plasticity, and enhancement of the GluN2B subunit expression enhances synaptic plasticity. For these reasons, allosteric potentiators of the GluN2B subunit have long been predicted to act as cognitive enhancers, with potential clinical utility in a wide range of conditions including Alzheimer's disease, post-injury motor learning, and schizophrenia. Work supported by the previous funding cycle allowed us to define the structural determinants and mechanism of action of 2 new classes of negative allosteric NMDA receptor modulators and 3 new classes of allosteric potentiators that are highly selective for NMDA receptors containing the GluN2C or GluN2C/D subunits. During the previous funding cycle, we also identified for the first time several drug-like GluN2B allosteric potentiators. The site and mechanism of action of GluN2B potentiation is distinct from that observed for GluN2C/D potentiators. Potentiation is absent when the alternatively spliced exon5 of GluN1 is included in the amino terminal domain, and these new compounds show strong potentiation (>5-fold) of responses to submaximal agonist responses in part due to allosteric enhancement of glutamate and/or glycine potency. These GluN2B potentiators do not act at any known modulator binding site, and thus break new ground in terms of NMDA receptor regulation. We propose 3 experiments that will advance our understanding of NMDA receptor gating, the site and mechanism of action of these modulators, the role of GluN2B in synaptic plasticity, and the effects of GluN2B potentiation in learning/memory.
Aim 1 : What is the mechanism of action of GluN2B potentiation? We will use whole cell voltage clamp and single channel recordings from recombinant and neuronal GluN2B-containing NMDA receptors to elucidate the mechanism by which representative members of two classes of compounds potentiate receptor function.
Aim 2 : What are the structural determinants of GluN2B potentiators? We will utilize a chimeric strategy and mutagenesis through of preM1/M1 regions in GluN1 and GluN2B to identify the structural determinants of action. We have developed azide and benzophenone photoaffinity labels that are active and should covalently label residues near the binding pocket of His-tagged GluN1 and GluN2 subunits.
Aim 3 : Does potentiation of GluN2B function alter synaptic plasticity? We will assess the ability of GluN2B potentiation to shift the threshold necessary to induce plasticit (LTP, LTD) in hippocampal slices.
Aim 4 : Does potentiation of GluN2B function alter learning and memory in vivo? We will assess in vivo the effects on learning for the class of GluN2B allosteric potentiator that is brain permeable.

Public Health Relevance

NMDA receptors, which are comprised of GluN1 and GluN2 subunits, mediate communication between neurons and thus play an important role in brain function and neurological diseases. Although potentiators and inhibitors selective for each of the four GluN2 subunits (A-D) have been proposed as useful therapeutic agents, subunit-selective inhibitors only exist for GluN2B (ifenprodil) and more recently GluN2A (TCN201). Work completed during the previous funding cycle (2009-2013), led to the first description of potent and subunit-selective GluN2C/D modulators and also revealed a novel class of compounds that selectively potentiate GluN2B. We will determine the mechanisms and effects of this new class of subunit-selective potentiators, which provide the first opportunity to test whether GluN2B potentiation could be useful for cognitive enhancement, memory loss, schizophrenia, and other health problems.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
4R01NS065371-08
Application #
9112009
Study Section
Neurotransporters, Receptors, and Calcium Signaling Study Section (NTRC)
Program Officer
Silberberg, Shai D
Project Start
2009-04-01
Project End
2018-07-31
Budget Start
2016-08-01
Budget End
2017-07-31
Support Year
8
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Emory University
Department
Pharmacology
Type
Schools of Medicine
DUNS #
066469933
City
Atlanta
State
GA
Country
United States
Zip Code
30322
Singh, Arun; Jenkins, Meagan A; Burke Jr, Kenneth J et al. (2018) Glutamatergic Tuning of Hyperactive Striatal Projection Neurons Controls the Motor Response to Dopamine Replacement in Parkinsonian Primates. Cell Rep 22:941-952
Bhattacharya, Subhrajit; Khatri, Alpa; Swanger, Sharon A et al. (2018) Triheteromeric GluN1/GluN2A/GluN2C NMDARs with Unique Single-Channel Properties Are the Dominant Receptor Population in Cerebellar Granule Cells. Neuron 99:315-328.e5
Fry, Andrew E; Fawcett, Katherine A; Zelnik, Nathanel et al. (2018) De novo mutations in GRIN1 cause extensive bilateral polymicrogyria. Brain :
Hansen, Kasper B; Yi, Feng; Perszyk, Riley E et al. (2018) Structure, function, and allosteric modulation of NMDA receptors. J Gen Physiol 150:1081-1105
Bhattacharya, Subhrajit; Ma, Yuxian; Dunn, Amy R et al. (2018) NMDA receptor blockade ameliorates abnormalities of spike firing of subthalamic nucleus neurons in a parkinsonian nonhuman primate. J Neurosci Res 96:1324-1335
Swanger, Sharon A; Vance, Katie M; Acker, Timothy M et al. (2018) A Novel Negative Allosteric Modulator Selective for GluN2C/2D-Containing NMDA Receptors Inhibits Synaptic Transmission in Hippocampal Interneurons. ACS Chem Neurosci 9:306-319
Fernández-Marmiesse, Ana; Kusumoto, Hirofumi; Rekarte, Saray et al. (2018) A novel missense mutation in GRIN2A causes a nonepileptic neurodevelopmental disorder. Mov Disord 33:992-999
Kaiser, Thomas M; Kell, Steven A; Kusumoto, Hirofumi et al. (2018) The Bioactive Protein-Ligand Conformation of GluN2C-Selective Positive Allosteric Modulators Bound to the NMDA Receptor. Mol Pharmacol 93:141-156
Perszyk, Riley; Katzman, Brooke M; Kusumoto, Hirofumi et al. (2018) An NMDAR positive and negative allosteric modulator series share a binding site and are interconverted by methyl groups. Elife 7:
Bhattacharya, Subhrajit; Traynelis, Stephen F (2018) Unique Biology and Single-Channel Properties of GluN2A- and GluN2C-Containing Triheteromeric N-Methyl-D-Aspartate Receptors. J Exp Neurosci 12:1179069518810423

Showing the most recent 10 out of 48 publications