UB Vice President for Research and Economic Development markup
Gabriela K. Popescu UB Profile
Biochemistry
Professor
955 Main Street
Buffalo NY 14203
Office 4226; Lab 4250
Buffalo NY 14203
716-829-3807
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Gary J. Iacobucci, Gabriela K. Popescu
Journal-article: The Journal of Neuroscience, vol. 39, pp. 8831-8844, 2019
Authors: Gary J. Iacobucci, Gabriela K. Popescu
Reference-entry: The Oxford Handbook of Neuronal Ion Channels, 2019
Authors: Gary J. Iacobucci, Gabriela K. Popescu
Journal-article: Biophysical Journal, 2019
Authors: Gary J. Iacobucci, Han Wen, Matthew Helou, Wenjun Zheng, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 116, pp. 106a, 2019
Authors: Gary J. Iacobucci, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 114, pp. 125a, 2018
Authors: Gary J Iacobucci, Gabriela K Popescu
Journal-article: Current Opinion in Physiology, vol. 2, pp. 114-122, 2018
Authors: Wenjun Zheng, Han Wen, Gary J. Iacobucci, Gabriela K. Popescu
Abstract: N-Methyl-D-aspartate (NMDA) receptors are glutamate-gated excitatory channels that play essential roles in brain functions. High-resolution structures have been solved for an allosterically inhibited and agonist-bound form of a functional NMDA receptor; however, other key functional states (particularly the active open-channel state) were only resolved at moderate resolutions by cryo-electron microscopy (cryo-EM). To decrypt the mechanism of the NMDA receptor activation, structural modeling is essential to provide presently missing information about structural dynamics. We performed systematic coarse-grained modeling using an elastic network model and related modeling/analysis tools (e.g., normal mode analysis, flexibility and hotspot analysis, cryo-EM flexible fitting, and transition pathway modeling) based on an active-state cryo-EM map. We observed extensive conformational changes that allosterically couple the extracellular regulatory and agonist-binding domains to the pore-forming trans-membrane domain (TMD), and validated these, to our knowledge, new observations against known mutational and functional studies. Our results predict two key modes of collective motions featuring shearing/twisting of the extracellular domains relative to the TMD, reveal subunit-specific flexibility profiles, and identify functional hotspot residues at key domain-domain interfaces. Finally, by examining the conformational transition pathway between the allosterically inhibited form and the active form, we predict a discrete sequence of domain motions, which propagate from the extracellular domains to the TMD. In summary, our results offer rich structural and dynamic information, which is consistent with the literature on structure-function relationships in NMDA receptors, and will guide in-depth studies on the activation dynamics of this important neurotransmitter receptor.
PubMedId: 28636915
Journal-article: Biophysical Journal, vol. 112, pp. 2589-2601, 2017
Authors: Vivek K. Bajpai, Laura Kerosuo, Georgios Tseropoulos, Kirstie A. Cummings, Xiaoyan Wang, Pedro Lei, Biao Liu, Song Liu, Gabriela K. Popescu, Marianne E. Bronner, Stelios T. Andreadis
Keywords: Epidermal keratinocytes Neural crest Neural crest induction Neural plate border Reprogramming
Abstract: During development, neural crest (NC) cells are induced by signaling events at the neural plate border of all vertebrate embryos. Initially arising within the central nervous system, NC cells subsequently undergo an epithelial to mesenchymal transition to migrate into the periphery, where they differentiate into diverse cell types. Here we provide evidence that postnatal human epidermal keratinocytes (KC), in response to fibroblast growth factor 2 and insulin like growth factor 1 signals, can be reprogrammed toward a NC fate. Genome-wide transcriptome analyses show that keratinocyte-derived NC cells are similar to those derived from human embryonic stem cells. Moreover, they give rise in vitro and in vivo to NC derivatives such as peripheral neurons, melanocytes, Schwann cells and mesenchymal cells (osteocytes, chondrocytes, adipocytes, and smooth muscle cells). By demonstrating that human keratin-14+ KC can form NC cells, even from clones of single cells, our results have important implications in stem cell biology and regenerative medicine. Stem Cells 2017;35:1402-1415.
PubMedId: 28142205
Journal-article: STEM CELLS, vol. 35, pp. 1402-1415, 2017
Authors: Gary J. Iacobucci, Gabriela K. Popescu
Journal-article: Nature Reviews Neuroscience, vol. 18, pp. 236-249, 2017
Authors: Gary J Iacobucci, Gabriela K Popescu
Abstract: N-methyl-d-aspartate (NMDA) receptors are glutamate- and glycine-gated channels that flux Na+ and Ca2+ into postsynaptic neurons during synaptic transmission. The resulting intracellular Ca2+ transient is essential to physiological and pathological processes related to synaptic development, plasticity, and apoptosis. It also engages calmodulin (CaM) to reduce subsequent NMDA receptor activity in a process known as Ca2+-dependent inactivation (CDI). Here, we used whole-cell electrophysiology to measure CDI and computational modeling to dissect the sequence of events that underlies it. With these approaches, we estimate that CaM senses NMDA receptor Ca2+ influx at 9nm from the channel pore. Further, when we controlled the frequency of Ca2+ influx through individual channels, we found that a kinetic model where apoCaM associates with channels before their activation best predicts the measured CDI. These results provide, to our knowledge, novel functional evidence for CaM preassociation to NMDA receptors in living cells. This particular mechanism for autoinhibitory feedback reveals strategies and challenges for Ca2+ regulation in neurons during physiological synaptic activity and disease.
PubMedId: 28712640
Journal-article: Biophysical journal, vol. 113, 2017
Authors: Mohammad Mehdi Maneshi, Bruce Maki, Radhakrishnan Gnanasambandam, Sophie Belin, Gabriela K. Popescu, Frederick Sachs, Susan Z. Hua
Abstract: While studying the physiological response of primary rat astrocytes to fluid shear stress in a model of traumatic brain injury (TBI), we found that shear stress induced Ca2+ entry. The influx was inhibited by MK-801, a specific pore blocker of N-Methyl-D-aspartic acid receptor (NMDAR) channels, and this occurred in the absence of agonists. Other NMDA open channel blockers ketamine and memantine showed a similar effect. The competitive glutamate antagonists AP5 and GluN2B-selective inhibitor ifenprodil reduced NMDA-activated currents, but had no effect on the mechanically induced Ca2+ influx. Extracellular Mg2+ at 2mM did not significantly affect the shear induced Ca2+ influx, but at 10mM it produced significant inhibition. Patch clamp experiments showed mechanical activation of NMDAR and inhibition by MK-801. The mechanical sensitivity of NMDARs may play a role in the normal physiology of fluid flow in the glymphatic system and it has obvious relevance to TBI.
PubMedId: 28045032
Journal-article: Scientific Reports, vol. 7, pp. 39610, 2017
Authors: Kirstie A. Cummings, Gary J. Iacobucci, Gabriela K. Popescu
Book-chapter: Ionotropic Glutamate Receptor Technologies, pp. 273-299, 2016
Authors: Kirstie A Cummings, Sophie Belin, Gabriela K Popescu
Keywords: Glycine Kinetics N-methyl-d-aspartate receptors Splice variants Whole-cell electrophysiology Zinc pH
Abstract: N-methyl-d-aspartate (NMDA) receptors assembled from GluN1 and GluN3 subunits are unique in that they form glycine-gated excitatory channels that are insensitive to glutamate and NMDA. Alternative splicing of the GluN1 subunit mRNA results in eight variants with regulated expression patterns and post-translational modifications. Here we investigate the role of residues in the GluN1 C-terminal alternatively spliced cassettes in receptor gating and modulation. We measured whole-cell currents from recombinant GluN1/GluN3A receptors expressed in HEK293cells that differed in the sequence of their GluN1 C-terminal tail. We found that these residues controlled the level of steady-state activity and the degree to which activity was facilitated by zinc and protons. Further, we found that the phosphorylation status of sites specific to certain variants can also modulate channel activity. Based on these results we suggest that GluN1 C-terminal domain splicing may confer cell-specific and activity-dependent regulation onto the level and pharmacologic sensitivity of GluN1/GluN3A currents.
PubMedId: 28365212
Journal-article: Neuropharmacology, vol. 119, 2016
Authors: Aurelian Buz??ianu, Ioana Cs??ki, Petra Mo??oiu, Gabriela Popescu, Ingolfur Thorbjornsson, Kolbrun R. Ragnarsodottir, S??mundur Gu??laugsson, Daniel Goubmunson
Journal-article: Advanced Materials Research, vol. 1114, pp. 233-238, 2015
Authors: Kirstie A Cummings, Gabriela K Popescu
Abstract: N-methyl-D-aspartate (NMDA) receptors are glutamate- and glycine-gated channels composed of two GluN1 and two GluN2 or/and GluN3 subunits. GluN3A expression is developmentally regulated, and changes in this normal pattern of expression, which occur in several brain disorders, alter synaptic maturation and function by unknown mechanisms. Uniquely within the NMDA receptor family, GluN1/GluN3 receptors produce glycine-gated deeply desensitising currents that are insensitive to glutamate and NMDA; these currents remain poorly characterised and their cellular functions are unknown. Here, we show that extracellular acidification strongly potentiated glycine-gated currents from recombinant GluN1/GluN3A receptors, with half-maximal effect in the physiologic pH range. This was largely due to slower current desensitisation and faster current recovery from desensitisation, and was mediated by residues facing the heterodimer interface of the ligand-binding domain. Consistent with the observed changes in desensitisation kinetics, acidic shifts increased the GluN1/GluN3A equilibrium current and depolarized the membrane in a glycine concentration-dependent manner. These results reveal novel modulatory mechanisms for GluN1/GluN3A receptors that further differentiate them from the canonical glutamatergic GluN1/GluN2 receptors and provide a new and potent pharmacologic tool to assist the detection, identification, and the further study of GluN1/GluN3A currents in native preparations.
PubMedId: 27000430
Journal-article: Scientific reports, vol. 6, 2015
Authors: William F Borschel, Kirstie A Cummings, LeeAnn K Tindell, Gabriela K Popescu
Keywords: N-methyl-d-aspartate receptor (NMDA receptor, NMDAR) activation mechanism gating ionotropic glutamate receptor kinetics receptor structure-function synaptic transmission
Abstract: Among glutamate-gated channels, NMDA receptors produce currents that subside with unusually slow kinetics, and this feature is essential to the physiology of central excitatory synapses. Relative to the homologous AMPA and kainate receptors, NMDA receptors have additional intersubunit contacts in the ligand binding domain that occur at both conserved and non-conserved sites. We examined GluN1/GluN2A single-channel currents with kinetic analyses and modeling to probe these class-specific intersubunit interactions for their role in glutamate binding and receptor gating. We found that substitutions that eliminate such interactions at non-conserved sites reduced stationary gating, accelerated deactivation, and imparted sensitivity to aniracetam, an AMPA receptor-selective positive modulator. Abolishing unique contacts at conserved sites also reduced stationary gating and accelerated deactivation. These results show that contacts specific to NMDA receptors, which brace the heterodimer interface within the ligand binding domain, stabilize actively gating receptor conformations and result in longer bursts and slower deactivations. They support the view that the strength of the heterodimer interface modulates gating in both NMDA and non-NMDA receptors and that unique interactions at this interface are responsible in part for basic differences between the kinetics of NMDA and non-NMDA currents at glutamatergic synapses.
PubMedId: 26370091
Journal-article: The Journal of biological chemistry, vol. 290, 2015
Authors: Meaghan A Paganelli, Gabriela K Popescu
Keywords: NMDA electrophysiology local anesthetics pain single-channel
Abstract: NMDA receptors mediate excitatory neurotransmission in brain and spinal cord and play a pivotal role in the neurological disease state of chronic pain, which is caused by central sensitization. Bupivacaine is the indicated local anesthetic in caudal, epidural, and spinal anesthesia and is widely used clinically to manage acute and chronic pain. In addition to blocking Na(+) channels, bupivacaine affects the activity of many other channels, including NMDA receptors. Importantly, bupivacaine inhibits NMDA receptor-mediated synaptic transmission in the dorsal horn of the spinal cord, an area critically involved in central sensitization. We used recombinant NMDA receptors expressed in HEK293 cells and found that increasing concentrations of bupivacaine decreased channel open probability in GluN2 subunit- and pH-independent manner by increasing the mean duration of closures and decreasing the mean duration of openings. Using kinetic modeling of one-channel currents, we attributed the observed current decrease to two main mechanisms: a voltage-dependent "foot-in-the-door" pore block and an allosteric gating effect. Further, the inhibition was state-independent because it occurred to the same degree whether the drug was applied before or after glutamate stimulation and was mediated by extracellular and intracellular inhibitory sites, via hydrophilic and hydrophobic pathways. These results predict that clinical doses of bupivacaine would decrease the peak and accelerate the decay of synaptic NMDA receptor currents during normal synaptic transmission. These quantitative predictions inform possible applications of bupivacaine as preventative and therapeutic approaches in chronic pain.
PubMedId: 25589775
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 35, 2015
Authors: Gary J. Iacobucci, Bruce A. Maki, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 108, pp. 285a, 2015
Authors: Kirstie A. Cummings, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 108, pp. 285a, 2015
Authors: Kirstie A Cummings, Gabriela K Popescu
Abstract: N-methyl-d-aspartate (NMDA) receptors are the only neurotransmitter receptors whose activation requires two distinct agonists. Heterotetramers of two GluN1 and two GluN2 subunits, NMDA receptors are broadly distributed in the central nervous system, where they mediate excitatory currents in response to synaptic glutamate release. Pore opening depends on the concurrent presence of glycine, which modulates the amplitude and time course of the glutamate-elicited response. Gating schemes for fully glutamate- and glycine-bound NMDA receptors have been described in sufficient detail to bridge the gap between microscopic and macroscopic receptor behaviors; for several receptor isoforms, these schemes include glutamate-binding steps. We examined currents recorded from cell-attached patches containing one GluN1/GluN2A receptor in the presence of several glycine-site agonists and used kinetic modeling of these data to develop reaction schemes that include explicit glycine-binding steps. Based on the ability to match a series of experimentally observed macroscopic behaviors, we propose a model for activation of the glutamate-bound NMDA receptor by glycine that predicts apparent negative agonist cooperativity and glycine-dependent desensitization in the absence of changes in microscopic binding or desensitization rate constants. These results complete the basic steps of an NMDA receptor reaction scheme for the GluN1/GluN2A isoform and prompt a reevaluation of how glycine controls NMDA receptor activation. We anticipate that our model will provide a useful quantitative instrument to further probe mechanisms and structure-function relationships of NMDA receptors and to better understand the physiological and pathological implications of endogenous fluctuations in extracellular glycine concentrations.
PubMedId: 25964432
Journal-article: The Journal of general physiology, vol. 145, 2014
Authors: Bruce A Maki, Gabriela K Popescu
Abstract: Brief intracellular Ca(2+) transients initiate signaling routines that direct cellular activities. Consequently, activation of Ca(2+)-permeable neurotransmitter-gated channels can both depolarize and initiate remodeling of the postsynaptic cell. In particular, the Ca(2+) transient produced by NMDA receptors is essential to normal synaptic physiology, drives the development and plasticity of excitatory central synapses, and also mediates glutamate excitotoxicity. The amplitude and time course of the Ca(2+) signal depends on the receptor's conductance and gating kinetics; these properties are themselves influenced both directly and indirectly by fluctuations in the extracellular Ca(2+) concentration. Here, we used electrophysiology and kinetic modeling to delineate the direct effects of extracellular Ca(2+) on recombinant GluN1/GluN2A receptor conductance and gating. We report that, in addition to decreasing unitary conductance, Ca(2+) also decreased channel open probability primarily by lengthening closed-channel periods. Using one-channel current recordings, we derive a kinetic model for GluN1/GluN2A receptors in physiological Ca(2+) concentrations that accurately describes macroscopic channel behaviors. This model represents a practical instrument to probe the mechanisms that control the Ca(2+) transients produced by NMDA receptors during both normal and aberrant synaptic signaling.
PubMedId: 25348411
Journal-article: The Journal of general physiology, vol. 144, 2014
Authors: Teresa K Aman, Bruce A Maki, Thomas J Ruffino, Eileen M Kasperek, Gabriela K Popescu
Keywords: Calcium Signaling Ionotropic Glutamate Receptor Neurotransmitter Receptor Permeation Mechanism Protein Phosphorylation Reaction Mechanism Receptor Desensitization Single-channel Kinetics cAMP Protein Kinase
Abstract: Protein kinase A (PKA) enhances synaptic plasticity in the central nervous system by increasing NMDA receptor current amplitude and Ca(2+) flux in an isoform-dependent yet poorly understood manner. PKA phosphorylates multiple residues on GluN1, GluN2A, and GluN2B subunits in vivo, but the functional significance of this multiplicity is unknown. We examined gating and permeation properties of recombinant NMDA receptor isoforms and of receptors with altered C-terminal domain (CTDs) prior to and after pharmacological inhibition of PKA. We found that PKA inhibition decreased GluN1/GluN2B but not GluN1/GluN2A gating; this effect was due to slower rates for receptor activation and resensitization and was mediated exclusively by the GluN2B CTD. In contrast, PKA inhibition reduced NMDA receptor-relative Ca(2+) permeability (PCa/PNa) regardless of the GluN2 isoform and required the GluN1 CTD; this effect was due primarily to decreased unitary Ca(2+) conductance, because neither Na(+) conductance nor Ca(2+)-dependent block was altered substantially. Finally, we show that both the gating and permeation effects can be reproduced by changing the phosphorylation state of a single residue: GluN2B Ser-1166 and GluN1 Ser-897, respectively. We conclude that PKA effects on NMDA receptor gating and Ca(2+) permeability rely on distinct phosphorylation sites located on the CTD of GluN2B and GluN1 subunits. This separate control of NMDA receptor properties by PKA may account for the specific effects of PKA on plasticity during synaptic development and may lead to drugs targeted to alter NMDA receptor gating or Ca(2+) permeability.
PubMedId: 24847051
Journal-article: The Journal of biological chemistry, vol. 289, 2014
Authors: Gabriela K Popescu
PubMedId: 25512600
Journal-article: The Journal of general physiology, vol. 145, 2014
Authors: Jessica A. Murphy, Ivar S. Stein, C. Geoffrey Lau, Rui T. Peixoto, Teresa K. Aman, Naoki Kaneko, Kelly Aromolaran, Jessica L. Saulnier, Gabriela K. Popescu, Bernardo L. Sabatini, Johannes W. Hell, R. Suzanne Zukin
Abstract: The NMDA-type glutamate receptor (NMDAR) is essential for synaptogenesis, synaptic plasticity, and higher cognitive function. Emerging evidence indicates that NMDAR Ca(2+) permeability is under the control of cAMP/protein kinase A (PKA) signaling. Whereas the functional impact of PKA on NMDAR-dependent Ca(2+) signaling is well established, the molecular target remains unknown. Here we identify serine residue 1166 (Ser1166) in the carboxy-terminal tail of the NMDAR subunit GluN2B to be a direct molecular and functional target of PKA phosphorylation critical to NMDAR-dependent Ca(2+) permeation and Ca(2+) signaling in spines. Activation of -adrenergic and D1/D5-dopamine receptors induces Ser1166 phosphorylation. Loss of this single phosphorylation site abolishes PKA-dependent potentiation of NMDAR Ca(2+) permeation, synaptic currents, and Ca(2+) rises in dendritic spines. We further show that adverse experience in the form of forced swim, but not exposure to fox urine, elicits striking phosphorylation of Ser1166 in vivo, indicating differential impact of different forms of stress. Our data identify a novel molecular and functional target of PKA essential to NMDAR-mediated Ca(2+) signaling at synapses and regulated by the emotional response to stress.
PubMedId: 24431445
Journal-article: The Journal of Neuroscience, vol. 34, pp. 869-879, 2014
Authors: Bruce A. Maki, Kirstie A. Cummings, Meaghan A. Paganelli, Swetha E. Murthy, Gabriela K. Popescu
Abstract: Ion channel proteins are universal devices for fast communication across biological membranes. The temporal signature of the ionic flux they generate depends on properties intrinsic to each channel protein as well as the mechanism by which it is generated and controlled and represents an important area of current research. Information about the operational dynamics of ion channel proteins can be obtained by observing long stretches of current produced by a single molecule. Described here is a protocol for obtaining one-channel cell-attached patch-clamp current recordings for a ligand gated ion channel, the NMDA receptor, expressed heterologously in HEK293 cells or natively in cortical neurons. Also provided are instructions on how to adapt the method to other ion channels of interest by presenting the example of the mechano-sensitive channel PIEZO1. This method can provide data regarding the channel's conductance properties and the temporal sequence of open-closed conformations that make up the channel's activation mechanism, thus helping to understand their functions in health and disease.
PubMedId: 24961614
Journal-article: Journal of Visualized Experiments, 2014
Authors: Han Yu, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 104, pp. 273a, 2013
Authors: Meaghan A. Paganelli, Cassandra L. Kussius, Gabriela K. Popescu
Abstract: In response to brief glutamate exposure, NMDA receptors produce excitatory currents that have sub-maximal amplitudes and characteristically slow kinetics. The activation sequence starts when glutamate binds to residues located on the upper lobe of extracellularly located ligand-binding domains (LBDs) and then contacts lower lobe residues to bridge the cleft between the two hinged lobes. This event stabilizes a narrow-cleft LBD conformation and may facilitate subsequent inter-lobe contacts that further stabilize the closed cleft. Agonist efficacy has been traced to the degree of agonist-induced cleft-closure and may also depend on the stability of the closed-cleft conformation. To investigate how cross-cleft contacts contribute to the amplitude and kinetics of NMDA receptor response, we examined the activation reaction of GluN1/GluN2A receptors that had single-residue substitutions at the interface between LBD lobes. We found that side-chain truncations at residues of putative contact between lobes increased glutamate efficacy through independent additive mechanisms in GluN1 and GluN2A subunits. In contrast, removing side-chain charge with isosteric substitutions at the same sites decreased glutamate efficacy. These results support the view that in GluN1/GluN2A receptors' natural interactions between residues on opposing sides of the ligand-binding cleft encode the stability of the glutamate-bound closed-cleft conformations and limit the degree of cleft closure, thus contributing to the sub-maximal response and emblematically slow NMDA receptor deactivation after brief stimulation.
PubMedId: 24278352
Journal-article: PLoS ONE, vol. 8, pp. e80953, 2013
Authors: Bruce A. Maki, Ross Cole, Gabriela K. Popescu
Keywords: NMDA receptors ionotropic glutamate receptors protein phosphorylation reaction mechanism single-channel kinetics
Abstract: NMDA receptors are glutamate-activated, Ca ( 2+) -permeable ion channels with critical roles in synaptic transmission and plasticity. The shape and size of their current is modulated by several kinase/phosphatase systems, and numerous residues located on the receptors' intracellular C-termini are phosphorylated in vivo. To investigate the mechanisms by which phosphorylation may control channel gating, we examined the single-channel behaviors of receptors carrying the S900A or S929A substitution in their GluN2A subunits and thus were rendered resistant to phosphorylation at those sites. We found that the mutations reduced channel open probability primarily by increasing the frequency of desensitized events. The kinetic models we developed revealed complex but similar changes in mechanism for the two mutants, leading to the view that dephosphorylation at either site may cause receptors to activate slower, deactivate faster and desensitize more frequently. This modulatory mechanism is consistent with the proposed roles for these residues in Ca ( 2+) -dependent desensitization and calcineurin-mediated reduction of current during brain development.
PubMedId: 23503100
Journal-article: Channels, vol. 7, pp. 126-132, 2013
Authors: H. Yu, G. K. Popescu
Abstract: N-Methyl-D-aspartate (NMDA) receptors mediate excitatory synaptic transmission in the central nervous system and play important roles in synaptic development and plasticity, but also mediate glutamate neurotoxicity. Recently, 2-naphthoic acid (NPA) and its derivatives have been identified as allosteric, noncompetitive NMDA receptor inhibitors. The selectivity of NPA derivatives among NMDA receptor subtypes was mapped structurally to the ligand-binding domain, and was proposed to be mediated by residues on the S1 segment. To delineate the kinetic mechanism by which NPA inhibits NMDA receptor activity, we examined its effects on the NMDA receptor gating reaction. Using whole-cell patch clamping on human embryonic kidney 293 cells expressing recombinant NMDA family of glutamate receptor subunits, GluN1/GluN2A, we found that NPA has a 50% inhibitory effect at 1.9 mM. Further, from one-channel current recordings, we found that 4 mM NPA caused a 62% decrease in open probability by decreasing mean open time 2.5-fold and by increasing mean closed time 2-fold. Kinetic modeling suggested that NPA binding stabilized NMDA receptor closed states and increased the energy barriers toward open states, causing NMDA receptors to dwell longer in pre-open states along the activation pathway. The reaction mechanisms we derived provide quantitative insight into the inhibitory mechanism of NPA and help anticipate its effects on GluN1/GluN2A receptors during both physiologic and pathologic activation modalities.
PubMedId: 23873856
Journal-article: Molecular Pharmacology, vol. 84, pp. 541-550, 2013
Authors: W. F. Borschel, J. M. Myers, E. M. Kasperek, T. P. Smith, N. M. Graziane, L. M. Nowak, G. K. Popescu
Abstract: The activation mechanisms of recombinant N-methyl-d-aspartate receptors (NRs) have been established in sufficient detail to account for their single channel and macroscopic responses; however, the reaction mechanism of native NRs remains uncertain due to indetermination of the isoforms expressed and possible neuron-specific factors. To delineate the activation mechanism of native NRs, we examined the kinetic properties of currents generated by individual channels located at the soma of cultured rat neurons. Cells were dissociated from the embryonic cerebral cortex or hippocampus, and on-cell single channel recordings were done between 4 and 50 days in vitro (DIV). We observed two types of kinetics that correlated with the age of the culture. When we segregated recordings by culture age, we found that receptors recorded from early (4-33 DIV) and late (25-50 DIV) cultures had smaller unitary conductances but had kinetic profiles that matched closely those of recombinant 2B- or 2A-containing receptors, respectively. In addition, we examined the effects of cotransfection with postsynaptic density protein 95 or neuropilin tolloid-like protein 1 on recombinant receptors expressed in human embryonic kidney-293 cells. Our results add support to the view that neuronal cultures recapitulate the developmental patterns of receptor expression observed in the intact animal and demonstrate that the activation mechanism of somatic neuronal NRs is similar to that described for recombinant receptors of defined subunit composition.
PubMedId: 22993263
Journal-article: Journal of Neurophysiology, vol. 108, pp. 3105-3115, 2012
Authors: Swetha E. Murthy, Tamer Shogan, Jessica C. Page, Eileen M. Kasperek, Gabriela K. Popescu
Abstract: N-methyl-D-aspartate (NMDA) receptor activation involves a dynamic series of structural rearrangements initiated by glutamate binding to glycine-loaded receptors and culminates with the clearing of the permeation pathway, which allows ionic flux. Along this sequence, three rate-limiting transitions can be quantified with kinetic analyses of single-channel currents, even though the structural determinants of these critical steps are unknown. In inactive receptors, the major permeation barrier resides at the intersection of four M3 transmembrane helices, two from each GluN1 and GluN2 subunits, at the level of the invariant SYTANLAAF sequence, known as the lurcher motif. Because the A7 but not A8 residues in this region display agonist-dependent accessibility to extracellular solutes, they were hypothesized to form the glutamate-sensitive gate. We tested this premise by examining the reaction mechanisms of receptors with substitutions in the lurcher motifs of GluN1 or GluN2A subunits. We found that, consistent with their locations relative to the proposed activation gate, A8Y decreased open-state stability, whereas A7Y dramatically stabilized open states, primarily by preventing gate closure; the equilibrium distribution of A7Y receptors was strongly shifted toward active states and resulted in slower microscopic association and dissociation rate constants for glutamate. In addition, for both A8- and A7-substituted receptors, we noticed patterns of kinetic changes that were specific to GluN1 or GluN2 locations. This may be a first indication that the sequence of discernible kinetic transitions during NMDA receptor activation may reflect subunit-dependent movements of M3 helices. Testing this hypothesis may afford insight into the activation mechanism of NMDA receptors.
PubMedId: 22891278
Journal-article: The Journal of General Physiology, vol. 140, pp. 267-277, 2012
Authors: S. A. Amico-Ruvio, M. A. Paganelli, J. M. Myers, G. K. Popescu
Abstract: N-Methyl-d-aspartate (NMDA) receptors are glutamate- and glycine-gated channels that mediate fast excitatory transmission in the central nervous system and are critical to synaptic development, plasticity, and integration. They have a rich complement of modulatory sites, which represent important pharmacological targets. Ifenprodil is a well tolerated NMDA receptor inhibitor; it is selective for GluN2B-containing receptors and has neuroprotective effects. The mechanism by which ifenprodil inhibits NMDA receptor responses is not fully understood. The inhibition is incomplete and noncompetitive with other known NMDA receptor agonists or modulators, although reciprocal effects have been reported between ifenprodil potency and that of extracellular ligands including glutamate, glycine, zinc, protons, and polyamines. Recent structural studies revealed that ifenprodil binds to a unique site at the interface between the extracellular N termini of GluN1 and GluN2B subunits, supporting the view that interactions with other extracellular modulators are indirect. In this study, we examined how ifenprodil affects the gating reaction of NMDA receptors in conditions designed to minimize actions by contemporaneous ligands. We found that ifenprodil decreased NMDA receptor equilibrium open probability by raising an energetic barrier to activation and also by biasing the receptor toward low open probability gating modes. These results demonstrate intrinsic effects of ifenprodil on NMDA receptor stationary gating kinetics and provide means to anticipate how ifenprodil will affect receptor responses in defined physiological and pathological circumstances.
PubMedId: 22936815
Journal-article: Molecular Pharmacology, vol. 82, pp. 1074-1081, 2012
Authors: Bruce A Maki, Teresa K Aman, Stacy A Amico-Ruvio, Cassandra L Kussius, Gabriela K Popescu
Abstract: NMDA receptors (NRs) are glutamate-gated calcium-permeable channels that are essential for normal synaptic transmssion and contribute to neurodegeneration. Tetrameric proteins consist of two obligatory GluN1 (N1) and two GluN2 (N2) subunits, of which GluN2A (2A) and GluN2B (2B) are prevalent in adult brain. The intracellularly located C-terminal domains (CTDs) make a significant portion of mass of the receptors and are essential for plasticity and excitotoxicity, but their functions are incompletely defined. Recent evidence shows that truncation of the N2 CTD alters channel kinetics; however, the mechanism by which this occurs is unclear. Here we recorded activity from individual NRs lacking the CTDs of N1, 2A, or 2B and determined the gating mechanisms of these receptors. Receptors lacking the N1 CTDs had larger unitary conductance and faster deactivation kinetics, receptors lacking the 2A or 2B CTDs had longer openings and longer desensitized intervals, and the first 100 amino acids of the N2 CTD were essential for these changes. In addition, receptors lacking the CTDs of either 2A or 2B maintained isoform-specific kinetic differences and swapping CTDs between 2A and 2B had no effect on single-channel properties. Based on these results, we suggest that perturbations in the CTD can modify the NR-mediated signal in a subunit-dependent manner, in 2A these effects are most likely mediated by membrane-proximal residues, and the isoform-specific biophysical properties conferred by 2A and 2B are CTD-independent. The kinetic mechanisms we developed afford a quantitative approach to understanding how the intracellular domains of NR subunits can modulate the responses of the receptor.
PubMedId: 22948148
Journal-article: The Journal of biological chemistry, vol. 287, 2012
Authors: Bruce E. Maki, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 100, pp. 269a-270a, 2011
Authors: William F. Borschel, Swetha E. Murthy, Eileen M. Kasperek, Gabriela K. Popescu
Abstract: Two classes of glutamate-activated channels mediate excitation at central synapses: N-methyl-D-aspartic acid (NMDA) receptors and non-NMDA receptors. Despite substantial structural homology, each class generates signals with characteristic kinetics and mediates distinct synaptic functions. In non-NMDA receptors, the strength of intersubunit contacts within ligand-binding domains is inversely correlated with functional desensitization. Here we test how the strength of these contacts affects NMDA receptor activation by combining mutagenesis and single-channel current analyses. We show that receptors with covalently linked dimers had significantly lower activity due to high barriers to opening and unstable open states but had intact desensitization. On the basis of these observations, we suggest that in NMDA receptors rearrangements at the heterodimer interface represent an early and integral step of the opening sequence but are not required for desensitization. These results demonstrate distinct functional roles in the activation of NMDA and non-NMDA glutamate-gated channels for largely conserved intersubunit contacts.
PubMedId: 21988914
Journal-article: Nature Communications, vol. 2, pp. 498, 2011
Authors: Gabriela K Popescu
Abstract: The time course of excitatory synaptic currents, the major means of fast communication between neurons of the central nervous system, is encoded in the dynamic behaviour of post-synaptic glutamate-activated channels. First-pass attempts to explain the glutamate-elicited currents with mathematical models produced reaction mechanisms that included only the most basic functionally defined states: resting vs. liganded, closed vs. open, responsive vs. desensitized. In contrast, single-molecule observations afforded by the patch-clamp technique revealed an unanticipated kinetic multiplicity of transitions: from microseconds-lasting flickers to minutes-long modes. How these kinetically defined events impact the shape of the synaptic response, how they relate to rearrangements in receptor structure, and whether and how they are physiologically controlled represent currently active research directions. Modal gating, which refers to the slowest, least frequently observed ion-channel transitions, has been demonstrated for representatives of all ion channel families. However, reaction schemes have been largely confined to the short- and medium-range time scales. For glutamate receptors as well, modal gating has only recently come under rigorous scrutiny. This article reviews the evidence for modal gating of glutamate receptors and the still developing hypotheses about the mechanism(s) by which modal shifts occur and the ways in which they may impact the time course of synaptic transmission.
PubMedId: 22106181
Journal-article: The Journal of physiology, vol. 590, 2011
Authors: Meaghan Paganelli, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 100, pp. 270a, 2011
Authors: Stacy??A. Amico-Ruvio, Swetha??E. Murthy, Thomas??P. Smith, Gabriela??K. Popescu
Abstract: Zinc accumulates in the synaptic vesicles of certain glutamatergic forebrain neurons and modulates neuronal excitability and synaptic plasticity by multiple poorly understood mechanisms. Zinc directly inhibits NMDA-sensitive glutamate-gated channels by two separate mechanisms: high-affinity binding to N-terminal domains of GluN2A subunits reduces channel open probability, and low-affinity voltage-dependent binding to pore-lining residues blocks the channel. Insight into the high-affinity allosteric effect has been hampered by the receptor's complex gating; multiple, sometimes coupled, modulatory mechanisms; and practical difficulties in avoiding transient block by residual Mg(2+). To sidestep these challenges, we examined how nanomolar zinc concentrations changed the gating kinetics of individual block-resistant receptors. We found that block-insensitive channels had lower intrinsic open probabilities but retained high sensitivity to zinc inhibition. Binding of zinc to these receptors resulted in longer closures and shorter openings within bursts of activity but had no effect on interburst intervals. Based on kinetic modeling of these data, we conclude that zinc-bound receptors have higher energy barriers to opening and less stable open states. We tested this model for its ability to predict zinc-dependent changes in macroscopic responses and to infer the impact of nanomolar zinc concentrations on synaptic currents mediated by 2A-type NMDA receptors.
PubMedId: 21504727
Journal-article: Biophysical Journal, vol. 100, pp. 1910-1918, 2011
Authors: Swetha Murthy, Jessica C. Page, Morgan E. Preziosi, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 100, pp. 24a, 2011
Authors: Gabriela K. Popescu, Swetha Murthy, William F. Borschel
Journal-article: Pharmaceuticals, vol. 3, pp. 3240-3257, 2010
Authors: William F. Borschel, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 98, pp. 524a-525a, 2010
Authors: Stacy A. Amico-Ruvio, Thomas P. Smith, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 98, pp. 526a, 2010
Authors: Cassandra L. Kussius, Andrei M. Popescu, Gabriela K. Popescu
Journal-article: Channels, vol. 4, pp. 78-82, 2010
Authors: Cassandra Kussius, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 98, pp. 525a, 2010
Authors: Swetha Murthy, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 98, pp. 525a, 2010
Authors: Stacy A. Amico-Ruvio, Gabriela K. Popescu
Journal-article: Biophysical Journal, vol. 98, pp. 1160-1169, 2010
Authors: Cassandra Kussius, Jason Myers, Kevin Barnum, Gabriela Popescu
Journal-article: Biophysical Journal, vol. 96, pp. 489a, 2009
Authors: Swetha Murthy, Gabriela Popescu
Journal-article: Biophysical Journal, vol. 96, pp. 490a, 2009
Authors: C. L. Kussius, N. Kaur, G. K. Popescu
Abstract: Neurosteroids are potent neuromodulators which act in part by binding to and modifying the activity of neurotransmitter-gated channels. Pregnanolone sulfate (PAS) is an endogenous neurosteroid that inhibits NMDA receptors and is neuroprotective in vivo. To delineate the mechanism of NMDA receptor inhibition by pregnanolone sulfate we used kinetic analyses of equilibrium single-channel currents recorded from individual GluN1/GluN2A receptors. Results show that PAS (0.1 mM) reduces single-channel open probability by 50% solely by increasing approximately 5-fold the mean time spent by receptors in closed conformations. From these data we derive a kinetic scheme that summarizes the effects of PAS on single channel kinetics, accounts for the PAS effects on macroscopic responses and leads us to propose that PAS inhibits NMDA receptor activity by shifting active receptors into desensitized conformations. These findings highlight the neurosteroid inhibitory site on NMDA receptors as a valuable therapeutic target against excitotoxic pathologies including acute and chronic neurodegeneration.
PubMedId: 19474309
Journal-article: Journal of Neuroscience, vol. 29, pp. 6819-6827, 2009
Authors: Cassandra L Kussius, Gabriela K Popescu
Abstract: Activation of ligand-gated channels is initiated by the binding of small molecules at extracellular sites and culminates with the opening of a membrane-embedded pore. To investigate how perturbations at ligand-binding domains influence the gating reaction, we examined current traces recorded from individual NMDA receptors in the presence of several subunit-specific partial agonists. We found that low-efficacy agonists acting at either the glycine-binding or the glutamate-binding NMDA receptor subunits had very similar effects on the receptor's activation reaction, possibly reflecting a high degree of coupling between the two subunit types during gating. In addition, we found that partial agonists increased the height of all energy barriers encountered by NMDA receptors during activation. This result stands in sharp contrast to the localized effects that have been observed for pentameric ligand-gated channels and may represent a previously unknown mechanism by which partial agonists reduce receptor activity.
PubMedId: 19648915
Journal-article: Nature Neuroscience, vol. 12, pp. 1114-1120, 2009
Authors: Stacy Amico, Navjot Kaur, Gabriela Popescu
Journal-article: Biophysical Journal, vol. 96, pp. 489a-490a, 2009
Authors: Jason Myers, William Borschel, Eileen Kasperek, Gabriela Popescu
Journal-article: Biophysical Journal, vol. 96, pp. 489a, 2009
Authors: Wei Zhang, James R Howe, Gabriela K Popescu
Abstract: Following brief stimulation, macroscopic NMDA receptor currents decay with biphasic kinetics that is believed to reflect glutamate dissociation and receptor desensitization. We found that the fast and slow decay components arise from the simultaneous deactivation of receptor populations that gate with short and long openings, respectively. Because individual receptors switched infrequently between gating modes, the relaxation time course was largely determined by the proportion of channels in each gating mode at the time of stimulation.
PubMedId: 18953348
Journal-article: Nature Neuroscience, vol. 11, pp. 1373-1375, 2008
Authors: G. Popescu
Journal-article: Cellular and Molecular Life Sciences, vol. 62, pp. 2100-2111, 2005
Authors: Gabriela Popescu
PubMedId: 16033953
1: Molecular pharmacology, vol. 68, 2005
Authors: Gabriela Popescu, Anthony Auerbach
Abstract: The slow component of the excitatory postsynaptic current in the central nervous system is generated by the activity of NMDA receptors. The activation properties of this class of glutamate receptor determine key features of the synaptic response and have important consequences for synaptic plasticity and cell physiology. NMDA receptor activation is complex and involves ligand binding, protein conformational changes, and channel blockade. Recently, two groups have proposed state models that encapsulate the essential features of NMDA receptor gating conformational changes. These models provide insight into the NMDA receptor as a molecular machine and should help us understand and manipulate NMDA receptor mediated synaptic function and pathologies.
PubMedId: 15155058
Journal-article: The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry, vol. 10, 2004
Authors: Gabriela Popescu, Antoine Robert, James R. Howe, Anthony Auerbach
Abstract: At central excitatory synapses, N-methyl-D-aspartate (NMDA) receptors, which have a high affinity for glutamate, produce a slowly rising synaptic current in response to a single transmitter pulse and an additional current after a second, closely timed stimulus. Here we show, by examining the kinetics of transmitter binding and channel gating in single-channel currents from recombinant NR1/NR2A receptors, that the synaptic response to trains of impulses is determined by the molecular reaction mechanism of the receptor. The rate constants estimated for the activation reaction predict that, after binding neurotransmitter, receptors hesitate for approximately 4 ms in a closed high-affinity conformation before they either proceed towards opening or release neurotransmitter, with about equal probabilities. Because only about half of the initially fully occupied receptors become active, repetitive stimulation elicits currents with distinct waveforms depending on pulse frequency. This high-affinity/low-efficiency activation mechanism might serve as a link between stimulation frequency and the directionality of the ensuing synaptic plasticity.
PubMedId: 15306812
Journal-article: Nature, vol. 430, pp. 790-793, 2004
Authors: Gabriela Popescu, Anthony Auerbach
PubMedId: 12679783
Journal-article: Nature Neuroscience, vol. 6, pp. 476-483, 2003
Authors: Hu Peng, Jason Myers, Xiaohong Fang, Ewa K Stachowiak, Pamela A Maher, Gabriel G Martins, Gabriela Popescu, Ronald Berezney, Michal K Stachowiak
PubMedId: 12065659
Journal Article: Journal of neurochemistry, vol. 81, 2002
Authors: Hu Peng, Jason Myers, Xiaohong Fang, Ewa K. Stachowiak, Pamela A. Maher, Gabriel G. Martins, Gabriela Popescu, Ronald Berezney???, Michal K. Stachowiak
Journal-article: Journal of Neurochemistry, vol. 81, pp. 506-524, 2002
Authors: Gabriela Popescu, Joseph L Napoli
Journal-article: Biochimica et Biophysica Acta (BBA) - Protein Structure and Molecular Enzymology, vol. 1476, pp. 43-52, 2000
Authors: G Popescu, J L Napoli
PubMedId: 10606766
Comparative Study: Biochimica et biophysica acta, vol. 1476, 1999
Authors: X Chai, Y Zhai, G Popescu, J L Napoli
PubMedId: 7499345
Comparative Study: The Journal of biological chemistry, vol. 270, 1995
Presented by CUBS © 2024