Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nora Lenkey is active.

Publication


Featured researches published by Nora Lenkey.


Molecular Pharmacology | 2006

The mechanism of activity-dependent sodium channel inhibition by the antidepressants fluoxetine and desipramine.

Nora Lenkey; Robert Karoly; Janos P. Kiss; Bernadett K. Szasz; E.S. Vizi; Arpad Mike

The effect of monoamine uptake inhibitor-type antidepressants on sodium channels of hippocampal neurons was investigated. Members of the tricyclic group of antidepressants are known to modify multiple targets, including sodium channels, whereas selective serotonin-reuptake inhibitors (SSRIs) are regarded as highly selective compounds, and their effect on sodium channels was not investigated in detail. In this study, a representative member of each group was chosen: the tricyclic antidepressant desipramine and the SSRI fluoxetine. The drugs were roughly equipotent use-dependent inhibitors of sodium channels, with IC50 values ∼100 μMat -150 mV holding potential, and ∼1 μMat -60 mV. We suggest that therapeutic concentrations of antidepressants affect neuronal information processing partly by direct, activity-dependent inhibition of sodium channels. As for the mechanism of inhibition, use-dependent inhibition by antidepressants was believed to be due to a preferential affinity to the fast-inactivated state. Using a voltage and perfusion protocol by which relative affinities to fast-versus slow-inactivated states could be assessed, we challenged this view and found that the affinity of both drugs to slowinactivated state(s) was higher. We propose a different mechanism of action for these antidepressants, in which slow rather than fast inactivation plays the dominant role. This mechanism is similar but not equivalent with the novel mechanism of usedependent sodium channel inhibition previously described by our group (Neuroscience 125:1019-1028, 2004; Neuroreport 14:1945-1949, 2003). Our results suggest that different drugs can produce use-dependent sodium channel inhibition by different mechanisms.


Neurochemistry International | 2012

GluN2B-containing NMDA receptors as possible targets for the neuroprotective and antidepressant effects of fluoxetine.

Janos P. Kiss; Bernadett K. Szasz; László Fodor; Arpad Mike; Nora Lenkey; Dalma Kurkó; József Nagy; E. Sylvester Vizi

Accumulating evidence has indicated the involvement of glutamatergic neurotransmission in the pathophysiology of excitotoxicity and in the mechanism of action of antidepressants. We have previously shown that tricyclic desipramine and the selective serotonin reuptake inhibitor fluoxetine inhibit NMDA receptors (NMDARs) in the clinically relevant, low micromolar concentration range. As the different subtypes of NMDARs are markedly different in their physiological and pathological functions, our aim was to investigate whether the effect of antidepressants is subtype-specific. Using whole-cell patch-clamp recordings in rat cortical cell cultures, we studied the age-dependence of inhibition of NMDA-induced currents after treatment with desipramine and fluoxetine, as the expression profile of the NMDAR subtypes changes as a function of days in vitro. We also investigated the inhibitory effect of these antidepressants on NMDA-induced currents in HEK 293 cell lines that stably expressed rat recombinant NMDARs with GluN1a/GluN2A or GluN1a/GluN2B subunit compositions. The inhibitory effect of desipramine was not age-dependent, whereas fluoxetine displayed a continuously decreasing inhibitory profile, which was similar to the GluN1/GluN2B subtype-selective antagonist ifenprodil. In HEK 293 cells, desipramine equally inhibited NMDA currents in both cell lines, whereas fluoxetine showed an inhibitory effect only in cells that expressed the GluN1/GluN2B subtype. Our data show that fluoxetine is a selective inhibitor of GluN2B-containing NMDARs, whereas desipramine inhibits both GluN1/GluN2A and GluN1/GluN2B subtypes. As the clinical efficacy of these drugs is very similar, the putative NMDAR-associated therapeutic effect of antidepressants may be mediated only via inhibition of the GluN2B-containing subtype. The manifestation of the GluN1/GluN2B-selectivity of fluoxetine suggests the neuroprotective potential for this drug in both acute and chronic neurodegenerative disorders.


PLOS Computational Biology | 2010

Fast- or Slow-inactivated State Preference of Na+ Channel Inhibitors: A Simulation and Experimental Study

Robert Karoly; Nora Lenkey; András Juhász; E. Sylvester Vizi; Arpad Mike

Sodium channels are one of the most intensively studied drug targets. Sodium channel inhibitors (e.g., local anesthetics, anticonvulsants, antiarrhythmics and analgesics) exert their effect by stabilizing an inactivated conformation of the channels. Besides the fast-inactivated conformation, sodium channels have several distinct slow-inactivated conformational states. Stabilization of a slow-inactivated state has been proposed to be advantageous for certain therapeutic applications. Special voltage protocols are used to evoke slow inactivation of sodium channels. It is assumed that efficacy of a drug in these protocols indicates slow-inactivated state preference. We tested this assumption in simulations using four prototypical drug inhibitory mechanisms (fast or slow-inactivated state preference, with either fast or slow binding kinetics) and a kinetic model for sodium channels. Unexpectedly, we found that efficacy in these protocols (e.g., a shift of the “steady-state slow inactivation curve”), was not a reliable indicator of slow-inactivated state preference. Slowly associating fast-inactivated state-preferring drugs were indistinguishable from slow-inactivated state-preferring drugs. On the other hand, fast- and slow-inactivated state-preferring drugs tended to preferentially affect onset and recovery, respectively. The robustness of these observations was verified: i) by performing a Monte Carlo study on the effects of randomly modifying model parameters, ii) by testing the same drugs in a fundamentally different model and iii) by an analysis of the effect of systematically changing drug-specific parameters. In patch clamp electrophysiology experiments we tested five sodium channel inhibitor drugs on native sodium channels of cultured hippocampal neurons. For lidocaine, phenytoin and carbamazepine our data indicate a preference for the fast-inactivated state, while the results for fluoxetine and desipramine are inconclusive. We suggest that conclusions based on voltage protocols that are used to detect slow-inactivated state preference are unreliable and should be re-evaluated.


PLOS ONE | 2010

Classification of drugs based on properties of sodium channel inhibition: a comparative automated patch-clamp study.

Nora Lenkey; Robert Karoly; Peter Lukacs; E. Sylvester Vizi; Morten Sunesen; Arpad Mike

Background There is only one established drug binding site on sodium channels. However, drug binding of sodium channels shows extreme promiscuity: ∼25% of investigated drugs have been found to potently inhibit sodium channels. The structural diversity of these molecules suggests that they may not share the binding site, and/or the mode of action. Our goal was to attempt classification of sodium channel inhibitors by measuring multiple properties of inhibition in electrophysiology experiments. We also aimed to investigate if different properties of inhibition correlate with specific chemical properties of the compounds. Methodology/Principal Findings A comparative electrophysiological study of 35 compounds, including classic sodium channel inhibitors (anticonvulsants, antiarrhythmics and local anesthetics), as well as antidepressants, antipsychotics and neuroprotective agents, was carried out using rNav1.2 expressing HEK-293 cells and the QPatch automatic patch-clamp instrument. In the multi-dimensional space defined by the eight properties of inhibition (resting and inactivated affinity, potency, reversibility, time constants of onset and offset, use-dependence and state-dependence), at least three distinct types of inhibition could be identified; these probably reflect distinct modes of action. The compounds were clustered similarly in the multi-dimensional space defined by relevant chemical properties, including measures of lipophilicity, aromaticity, molecular size, polarity and electric charge. Drugs of the same therapeutic indication typically belonged to the same type. We identified chemical properties, which were important in determining specific properties of inhibition. State-dependence correlated with lipophilicity, the ratio of the neutral form of molecules, and aromaticity: We noticed that the highly state dependent inhibitors had at least two aromatic rings, logP>4.0, and pKa<8.0. Conclusions/Significance The correlations of inhibition properties both with chemical properties and therapeutic profiles would not have been evident through the sole determination of IC50; therefore, recording multiple properties of inhibition may allow improved prediction of therapeutic usefulness.


The Journal of Neuroscience | 2014

Presynaptic Calcium Channel Inhibition Underlies CB1 Cannabinoid Receptor-Mediated Suppression of GABA Release.

Gergely G. Szabó; Nora Lenkey; Noemi Holderith; Tibor Andrási; Zoltan Nusser; Norbert Hájos

CB1 cannabinoid receptors (CB1) are located at axon terminals and effectively control synaptic communication and thereby circuit operation widespread in the CNS. Although it is partially uncovered how CB1 activation leads to the reduction of synaptic excitation, the mechanisms of the decrease of GABA release upon activation of these cannabinoid receptors remain elusive. To determine the mechanisms underlying the suppression of synaptic transmission by CB1 at GABAergic synapses, we recorded unitary IPSCs (uIPSCs) at cholecystokinin-expressing interneuron-pyramidal cell connections and imaged presynaptic [Ca2+] transients in mouse hippocampal slices. Our results reveal a power function with an exponent of 2.2 between the amplitude of uIPSCs and intrabouton [Ca2+]. Altering CB1 function by either increasing endocannabinoid production or removing its tonic activity allowed us to demonstrate that CB1 controls GABA release by inhibiting Ca2+ entry into presynaptic axon terminals via N-type (Cav2.2) Ca2+ channels. These results provide evidence for modulation of intrabouton Ca2+ influx into GABAergic axon terminals by CB1, leading to the effective suppression of synaptic inhibition.


Nature Communications | 2015

Tonic endocannabinoid-mediated modulation of GABA release is independent of the CB1 content of axon terminals

Nora Lenkey; Tekla Kirizs; Noemi Holderith; Zoltán Máté; Gábor Szabó; E. Sylvester Vizi; Norbert Hájos; Zoltan Nusser

The release of GABA from cholecystokinin-containing interneurons is modulated by type-1 cannabinoid receptors (CB1). Here we tested the hypothesis that the strength of CB1-mediated modulation of GABA release is related to the CB1 content of axon terminals. Basket cell boutons have on average 78% higher CB1 content than those of dendritic-layer-innervating (DLI) cells, a consequence of larger bouton surface and higher CB1 density. The CB1 antagonist AM251 caused a 54% increase in action potential-evoked [Ca2+] in boutons of basket cells, but not in DLI cells. However, the effect of AM251 did not correlate with CB1 immunoreactivity of individual boutons. Moreover, a CB1 agonist decreased [Ca2+] in a cell type- and CB1-content-independent manner. Replica immunogold labelling demonstrated the colocalization of CB1 with the Cav2.2 Ca2+ channel subunit. Our data suggest that only a subpopulation of CB1s, within nanometre distances from their target Cav2.2 channels, are responsible for endocannabinoid-mediated modulation of GABA release.


Planta Medica | 2008

Converging Effects of Ginkgo biloba Extract at the Level of Transmitter Release, NMDA and Sodium Currents and Dendritic Spikes

Bernadett K. Szasz; Nora Lenkey; Albert M. I. Barth; Arpad Mike; Zsolt Somogyvari; Orsolya Farkas; Balázs Lendvai

In this study, an attempt was made to integrate the effects of GINKGO BILOBA extract (GBE) in different experimental systems (IN VITRO cochlea, brain slice preparations and cortical cell culture) to elucidate whether these processes converge to promote neuroprotection or interfere with normal neural function. GBE increased the release of dopamine in the cochlea. NMDA-evoked currents were dose-dependently inhibited by rapid GBE application in cultured cortical cells. GBE moderately inhibited Na+ channels at depolarised holding potential in cortical cells. These inhibitory effects by GBE may sufficiently contribute to the prevention of excitotoxic damage in neurons. However, these channels also interact with memory formation at the cellular level. The lack of effect by GBE on dendritic spike initiation in neocortical layer 5 pyramidal neurons indicates that the integrative functions may remain intact during the inhibitory actions of GBE.


Frontiers in Pharmacology | 2015

Different pH-sensitivity patterns of 30 sodium channel inhibitors suggest chemically different pools along the access pathway

Alexandra A Lazar; Nora Lenkey; Krisztina Pesti; László Fodor; Arpad Mike

The major drug binding site of sodium channels is inaccessible from the extracellular side, drug molecules can only access it either from the membrane phase, or from the intracellular aqueous phase. For this reason, ligand-membrane interactions are as important determinants of inhibitor properties, as ligand-protein interactions. One-way to probe this is to modify the pH of the extracellular fluid, which alters the ratio of charged vs. uncharged forms of some compounds, thereby changing their interaction with the membrane. In this electrophysiology study we used three different pH values: 6.0, 7.3, and 8.6 to test the significance of the protonation-deprotonation equilibrium in drug access and affinity. We investigated drugs of several different indications: carbamazepine, lamotrigine, phenytoin, lidocaine, bupivacaine, mexiletine, flecainide, ranolazine, riluzole, memantine, ritanserin, tolperisone, silperisone, ambroxol, haloperidol, chlorpromazine, clozapine, fluoxetine, sertraline, paroxetine, amitriptyline, imipramine, desipramine, maprotiline, nisoxetine, mianserin, mirtazapine, venlafaxine, nefazodone, and trazodone. We recorded the pH-dependence of potency, reversibility, as well as onset/offset kinetics. As expected, we observed a strong correlation between the acidic dissociation constant (pKa) of drugs and the pH-dependence of their potency. Unexpectedly, however, the pH-dependence of reversibility or kinetics showed diverse patterns, not simple correlation. Our data are best explained by a model where drug molecules can be trapped in at least two chemically different environments: A hydrophilic trap (which may be the aqueous cavity within the inner vestibule), which favors polar and less lipophilic compounds, and a lipophilic trap (which may be the membrane phase itself, and/or lipophilic binding sites on the channel). Rescue from the hydrophilic and lipophilic traps can be promoted by alkalic and acidic extracellular pH, respectively.


Nature Communications | 2015

Erratum: Tonic endocannabinoid-mediated modulation of GABA release is independent of the CB 1 content of axon terminals (Nature Communications (2015) 6:6557 doi: 10.1038/ncomms7557)

Nora Lenkey; Tekla Kirizs; Noemi Holderith; Zoltán Máté; Gábor Szabó; E. Sylvester Vizi; Norbert Hájos; Zoltan Nusser

This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ ori.


Biophysical Journal | 2011

Physicochemical Properties of Sodium Channel Inhibitors which Determine Affinity to Resting and Depolarized States

Arpad Mike; Nora Lenkey; E. Sylvester Vizi

Affinity of inhibitor compounds to hyperpolarized and depolarized conformations of sodium channels (commonly termed resting and inactivated affinities: Kr and Ki, respectively) were investigated: Their correlations with chemical descriptors of inhibitors were studied, in order to deduce the nature of chemical interactions involved in binding to both conformations. Two separate approaches were used: (1) We performed a literature search, calculated Kr and Ki values, and created a database of altogether 204 Kr-Ki pairs obtained from 73 publications. (2) We carried out a comparative electrophysiological study of 35 drugs using rNav1.2 expressing HEK 293 cells and the QPatch automatic patch-clamp instrument.We observed that lipophilicity (quantified by the logarithm of the calculated water-octanol partition coefficient, logP) is important in determining both Kr and Ki, but had a greater effect on Ki. Distribution coefficients (logD) discriminated better between Kr and Ki than partition coefficients (logP). The ratio of positively charged/neutral forms (quantified by the acidic dissociation constant, pKa) was a significant determinant of resting affinity: predominantly charged compounds tended to be more potent against resting channels, while neutral compounds tended to be more state-dependent. Aromaticity was more important for inactivated state affinity.

Collaboration


Dive into the Nora Lenkey's collaboration.

Top Co-Authors

Avatar

Arpad Mike

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

E. Sylvester Vizi

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Robert Karoly

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Noemi Holderith

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Norbert Hájos

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Zoltan Nusser

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Bernadett K. Szasz

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

E.S. Vizi

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Tekla Kirizs

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Zoltán Máté

Hungarian Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge