Network


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

Hotspot


Dive into the research topics where Ilya Sukhanov is active.

Publication


Featured researches published by Ilya Sukhanov.


Neuropharmacology | 2005

Metabotropic glutamate receptor (mGluR5) antagonist MPEP attenuated cue- and schedule-induced reinstatement of nicotine self-administration behavior in rats.

Anton Bespalov; Olga A. Dravolina; Ilya Sukhanov; Elena Zakharova; Elena Blokhina; Edwin Zvartau; Wojciech Danysz; Gino Van Heeke; Athina Markou

Previous studies suggested that metabotropic glutamate 5 (mGlu5) receptors play an important role in the reinforcing effects of abused drugs. The present experiments evaluated the effects of the mGlu5 receptor antagonist, MPEP (2-methyl-6-(phenylethynyl)-pyridine hydrochloride; 1-10 mg/kg, salt, i.p.), in rat models of nicotine-seeking behavior that may have relevance to relapse to drug-taking. Male Wistar rats (with restricted access to food) were trained to nose-poke to receive intravenous infusions of nicotine (0.03 mg/kg per infusion, base) under a fixed ratio 5 time out 60 s schedule of reinforcement. After stable nicotine self-administration was acquired, nose-poking behavior was extinguished in the absence of nicotine-associated cues. During the reinstatement test phase, independent groups of animals were exposed to: (a) response-contingent nicotine-associated cues (cue-induced reinstatement); or (b) response-noncontingent presentations of 45-mg food pellets under fixed time 2 min schedule (schedule-induced reinstatement). Additional control experiments were conducted to demonstrate that in nicotine-naïve animals MPEP does not affect cue-induced reinstatement of food-seeking behavior and has no effects on operant behavior maintained by a simple fixed interval 2 min schedule of food reinforcement. Pretreatment with MPEP (10 mg/kg) significantly attenuated the reinstatement of nicotine-seeking in both experiments. Further, MPEP (10 mg/kg) significantly attenuated polydipsia induced by a fixed time 2 min food schedule. In conclusion, the present findings indicate that the blockade of mGlu5 receptors attenuates cue-induced reinstatement of nicotine self-administration behavior (but not food-seeking) and may produce a general inhibition of schedule-induced behaviors, including schedule-induced nicotine-seeking.


Neuropsychopharmacology | 2015

TAAR1 Modulates Cortical Glutamate NMDA Receptor Function

Stefano Espinoza; Gabriele Lignani; Lucia Caffino; Silvia Maggi; Ilya Sukhanov; Damiana Leo; Liudmila Mus; Marco Emanuele; Giuseppe Ronzitti; Anja Harmeier; Lucian Medrihan; Tatyana D. Sotnikova; Evelina Chieregatti; Marius C. Hoener; Fabio Benfenati; Valter Tucci; Fabio Fumagalli; Raul R. Gainetdinov

Trace Amine-Associated Receptor 1 (TAAR1) is a G protein-coupled receptor expressed in the mammalian brain and known to influence subcortical monoaminergic transmission. Monoamines, such as dopamine, also play an important role within the prefrontal cortex (PFC) circuitry, which is critically involved in high-o5rder cognitive processes. TAAR1-selective ligands have shown potential antipsychotic, antidepressant, and pro-cognitive effects in experimental animal models; however, it remains unclear whether TAAR1 can affect PFC-related processes and functions. In this study, we document a distinct pattern of expression of TAAR1 in the PFC, as well as altered subunit composition and deficient functionality of the glutamate N-methyl-D-aspartate (NMDA) receptors in the pyramidal neurons of layer V of PFC in mice lacking TAAR1. The dysregulated cortical glutamate transmission in TAAR1-KO mice was associated with aberrant behaviors in several tests, indicating a perseverative and impulsive phenotype of mutants. Conversely, pharmacological activation of TAAR1 with selective agonists reduced premature impulsive responses observed in the fixed-interval conditioning schedule in normal mice. Our study indicates that TAAR1 plays an important role in the modulation of NMDA receptor-mediated glutamate transmission in the PFC and related functions. Furthermore, these data suggest that the development of TAAR1-based drugs could provide a novel therapeutic approach for the treatment of disorders related to aberrant cortical functions.


Neuropharmacology | 2015

Postsynaptic D2 dopamine receptor supersensitivity in the striatum of mice lacking TAAR1

Stefano Espinoza; Valentina Ghisi; Marco Emanuele; Damiana Leo; Ilya Sukhanov; Tatiana D. Sotnikova; Evelina Chieregatti; Raul R. Gainetdinov

Trace Amine-Associated Receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) known to modulate dopaminergic system through several mechanisms. Mice lacking this receptor show a higher sensitivity to dopaminergic stimuli, such as amphetamine; however, it is not clear whether D1 or D2 dopamine receptors and which associated intracellular signaling events are involved in this modulation. In the striatum of TAAR1 knock out (TAAR1-KO mice) we found that D2, but not D1, dopamine receptors were over-expressed, both in terms of mRNA and protein levels. Moreover, the D2 dopamine receptor-related G protein-independent AKT/GSK3 signaling pathway was selectively activated, as indicated by the decrease of phosphorylation of AKT and GSK3β. The decrease in phospho-AKT levels, suggesting an increase in D2 dopamine receptor activity in basal conditions, was associated with an increase of AKT/PP2A complex, as revealed by co-immunoprecipitation experiments. Finally, we found that the locomotor activation induced by the D2 dopamine receptor agonist quinpirole, but not by the full D1 dopamine receptor agonist SKF-82958, was increased in TAAR1-KO mice. These data demonstrate pronounced supersensitivity of postsynaptic D2 dopamine receptors in the striatum of TAAR1-KO mice and indicate that a close interaction of TAAR1 and D2 dopamine receptors at the level of postsynaptic structures has important functional consequences.


Pharmacological Research | 2016

Increased context-dependent conditioning to amphetamine in mice lacking TAAR1

Ilya Sukhanov; Lucia Caffino; Evgeniya V. Efimova; Stefano Espinoza; Tatiana D. Sotnikova; Luigi Cervo; Fabio Fumagalli; Raul R. Gainetdinov

Given the recent evidence indicating that amphetamine derivatives may also act as direct agonists of the G protein-coupled trace amine-associated receptor 1 (TAAR1), we hypothesized that TAAR1 could contribute to the reinforcing and addictive properties of amphetamines. Accordingly, the present study aimed to investigate the role of TAAR1 in the effects of psychostimulants by analyzing context-dependent sensitization and conditioned place preference (CPP) to d-amphetamine (AMPH) in TAAR1-KO mice. In context-dependent sensitization experiment, TAAR1-KO mice showed higher conditioned locomotor responses compared to wild-type mice. In the CPP test, TAAR1-KO animals were also more sensitive to priming-induced reinstatement of AMPH-induced conditioned place preference (CPP) than wild type mice. Importantly, saline-treated and AMPH-treated mice lacking TAAR1 demonstrated significant alterations in the total levels and phosphorylation of the critical subunit of NMDA glutamate receptors, GluN1, in the striatum, suggesting a role of TAAR1 in the modulation of frontostriatal glutamate transmission; this effect could underlie the observed alterations in conditioning processes. In conclusion, our data suggest that TAAR1 receptors play an inhibitory role with respect to conditioned responses to AMPH by modulating, at least in part, corticostriatal glutamate transmission.


The Journal of Neuroscience | 2018

Pronounced Hyperactivity, Cognitive Dysfunctions, and BDNF Dysregulation in Dopamine Transporter Knock-out Rats

Damiana Leo; Ilya Sukhanov; Francesca Zoratto; Placido Illiano; Lucia Caffino; Fabrizio Sanna; Giulia Messa; Marco Emanuele; Alessandro Esposito; Mariia Dorofeikova; Evgeny A. Budygin; Liudmila Mus; Evgenia V. Efimova; Marco Niello; Stefano Espinoza; Tatyana D. Sotnikova; Marius C. Hoener; Giovanni Laviola; Fabio Fumagalli; Walter Adriani; Raul R. Gainetdinov

Dopamine (DA) controls many vital physiological functions and is critically involved in several neuropsychiatric disorders such as schizophrenia and attention deficit hyperactivity disorder. The major function of the plasma membrane dopamine transporter (DAT) is the rapid uptake of released DA into presynaptic nerve terminals leading to control of both the extracellular levels of DA and the intracellular stores of DA. Here, we present a newly developed strain of rats in which the gene encoding DAT knockout Rats (DAT-KO) has been disrupted by using zinc finger nuclease technology. Male and female DAT-KO rats develop normally but weigh less than heterozygote and wild-type rats and demonstrate pronounced spontaneous locomotor hyperactivity. While striatal extracellular DA lifetime and concentrations are significantly increased, the total tissue content of DA is markedly decreased demonstrating the key role of DAT in the control of DA neurotransmission. Hyperactivity of DAT-KO rats can be counteracted by amphetamine, methylphenidate, the partial Trace Amine-Associated Receptor 1 (TAAR1) agonist RO5203648 ((S)-4-(3,4-Dichloro-phenyl)-4,5-dihydro-oxazol-2-ylamine) and haloperidol. DAT-KO rats also demonstrate a deficit in working memory and sensorimotor gating tests, less propensity to develop obsessive behaviors and show strong dysregulation in frontostriatal BDNF function. DAT-KO rats could provide a novel translational model for human diseases involving aberrant DA function and/or mutations affecting DAT or related regulatory mechanisms. SIGNIFICANCE STATEMENT Here, we present a newly developed strain of rats in which the gene encoding the dopamine transporter (DAT) has been disrupted (Dopamine Transporter Knockout rats [DAT-KO rats]). DAT-KO rats display functional hyperdopaminergia accompanied by pronounced spontaneous locomotor hyperactivity. Hyperactivity of DAT-KO rats can be counteracted by amphetamine, methylphenidate, and a few other compounds exerting inhibitory action on dopamine-dependent hyperactivity. DAT-KO rats also demonstrate cognitive deficits in working memory and sensorimotor gating tests, less propensity to develop compulsive behaviors, and strong dysregulation in frontostriatal BDNF function. These observations highlight the key role of DAT in the control of brain dopaminergic transmission. DAT-KO rats could provide a novel translational model for human diseases involving aberrant dopamine functions.


Expert Opinion on Therapeutic Targets | 2018

Trace amine-associated receptor 1: a multimodal therapeutic target for neuropsychiatric diseases

Michael D. Schwartz; Juan J. Canales; Riccardo Zucchi; Stefano Espinoza; Ilya Sukhanov; Raul R. Gainetdinov

ABSTRACT Introduction: The trace amines, endogenous amines closely related to the biogenic amine neurotransmitters, have been known to exert physiological and neurological effects for decades. The recent identification of a trace amine-sensitive G protein-coupled receptor, trace amine-associated receptor 1 (TAAR1), and subsequent development of TAAR1-selective small-molecule ligands, has renewed research into the therapeutic possibilities of trace amine signaling. Areas covered: Recent efforts in elucidating the neuropharmacology of TAAR1, particularly in neuropsychiatric and neurodegenerative disease, addiction, and regulation of arousal state, will be discussed. Focused application of TAAR1 mutants, synthetic TAAR1 ligands, and endogenous biomolecules such as 3-iodothyronamine (T1AM) has yielded a basic functional portrait for TAAR1, despite a complex biochemistry and pharmacology. The close functional relationship between TAAR1 and dopaminergic signaling is likely to underlie many of its CNS effects. However, TAAR1’s influences on serotonin and glutamate neurotransmission will also be highlighted. Expert opinion: TAAR1 holds great promise as a therapeutic target for mental illness, addiction, and sleep disorders. A combination of preclinical and translationally driven studies has solidified TAAR1 as a key node in the regulation of dopaminergic signaling. Continued focus on the mechanisms underlying TAAR1’s regulation of serotonin and glutamate signaling, as well as dopamine, will yield further disease-relevant insights.


Neuropharmacology | 2018

Activation of trace amine-associated receptor 1 attenuates schedule-induced polydipsia in rats

Ilya Sukhanov; Artem Dorotenko; Antonina Dolgorukova; C. Marius Hoener; Raul R. Gainetdinov; Anton Bespalov Yu

&NA; Trace Amine Associated Receptor 1 (TAAR1) is a novel pharmacological target. TAAR1 are well‐documented to play a modulatory role in the dopaminergic system. In spite of a growing number of studies of TAAR1 effects, little is still known about the behavioral pharmacology of TAAR1 ligands, including effects of repeated TAAR1 agonist administration. The present study appears to be the first that estimated the action of TAAR1 agonists on schedule‐induced polydipsia, a type of adjunctive behavior, which is considered to be useful for evaluating certain aspects of obsessive‐compulsive and related disorders (OCD) and schizophrenia. Our results have demonstrated that the wide range of RO5263397, the highly selective partial TAAR1 agonist, doses (1–10 mg/kg) attenuated the polydipsia induced by two different schedules of food delivery in rats. The effect remained unchanged for the 7 days of repeated treatment. However, the highest tested doses of RO5263397 (6 and 10 mg/kg) decreased the vertical locomotor activity of the animals and the volume of water intake of thirsty rats following the acute treatment. Also, though, the repeated RO5263397 administration is exhibited to diminish the volume of consumed water and weight of rats without SIP, on the other hand, the tolerance was observed to these drug effects. In general, the RO5263397 decreases specifically the adjunctive drinking and this effect is maintained with repeated drug administration without the development of tolerance. The interpretation of these results as an evidence for the RO5263397 anticompulsive‐like action, however, should be taken with caution because the drug also influenced the drinking behavior and only weakly affected the other parameters of SIP used to reveal the potential anticompulsive‐like effects of drugs. HIGHLIGHTSRO5263397, the highly selective partial TAAR1 agonist, attenuated SIP in rats.This effect of the highest tested doses can be affected by non‐specific drug effects.The RO5263397 repeated administration did not influence the SIP‐attenuating effect.


Frontiers in Pharmacology | 2018

Behavioral Effects of a Potential Novel TAAR1 Antagonist

Vincent M. Lam; Catharine A. Mielnik; Corey Baimel; Pieter Beerepoot; Stefano Espinoza; Ilya Sukhanov; Wendy Horsfall; Raul R. Gainetdinov; Stephanie L. Borgland; Amy J. Ramsey; Ali Salahpour

The trace amine associated receptor 1 (TAAR1) is a G-protein coupled receptor expressed in the monoaminergic regions of the brain, and represents a potential novel therapeutic target for the treatment of neurological disorders. While selective agonists for TAAR1 have been successfully identified, only one high affinity TAAR1 antagonist has been described thus far. We previously identified four potential low potency TAAR1 antagonists through an in silico screen on a TAAR1 homology model. One of the identified antagonists (compound 22) was predicted to have favorable physicochemical properties, which would allow the drug to cross the blood brain barrier. In vivo studies were therefore carried out and showed that compound 22 potentiates amphetamine- and cocaine-mediated locomotor activity. Furthermore, electrophysiology experiments demonstrated that compound 22 increased firing of dopamine neurons similar to EPPTB, the only known TAAR1 antagonist. In order to assess whether the effects of compound 22 were mediated through TAAR1, experiments were carried out on TAAR1-KO mice. The results showed that compound 22 is able to enhance amphetamine- and cocaine-mediated locomotor activity, even in TAAR1-KO mice, suggesting that the in vivo effects of this compound are not mediated by TAAR1. In collaboration with Psychoactive Drug Screening Program, we attempted to determine the targets for compound 22. Psychoactive Drug Screening Program (PDSP) results suggested several potential targets for compound 22 including, the dopamine, norepinephrine and serotonin transporters; as well as sigma 1 and 2 receptors. Our follow-up studies using heterologous cell systems showed that the dopamine transporter is not a target of compound 22. Therefore, the biological target of compound 22 mediating its psychoactive effects still remains unknown.


Behavioural Brain Research | 2017

Novel reinforcement learning paradigm based on response patterning under interval schedules of reinforcement

Christin Schifani; Ilya Sukhanov; Mariia Dorofeikova; Anton Bespalov

HighlightsA novel operant reinforcement learning task for rats is presented.The task can be used repeatedly without any appreciable sign of a practice effect.The task is sensitive to cognitive deficits induced by NMDA receptor blockade. Abstract There is a need to develop cognitive tasks that address valid neuropsychological constructs implicated in disease mechanisms and can be used in animals and humans to guide novel drug discovery. Present experiments aimed to characterize a novel reinforcement learning task based on a classical operant behavioral phenomenon observed in multiple species – differences in response patterning under variable (VI) vs fixed interval (FI) schedules of reinforcement. Wistar rats were trained to press a lever for food under VI30 s and later weekly test sessions were introduced with reinforcement schedule switched to FI30 s. During the FI30 s test session, post‐reinforcement pauses (PRPs) gradually grew towards the end of the session reaching 22–43% of the initial values. Animals could be retrained under VI30 s conditions, and FI30 s test sessions were repeated over a period of several months without appreciable signs of a practice effect. Administration of the non‐competitive N‐methyl‐d‐aspartate (NMDA) receptor antagonist MK‐801 ((5S,10R)‐(+)‐5‐Methyl‐10,11‐dihydro‐5H‐dibenzo[a,d]cyclohepten‐5,10‐imine maleate) prior to FI30 s sessions prevented adjustment of PRPs associated with the change from VI to FI schedule. This effect was most pronounced at the highest tested dose of MK‐801 and appeared to be independent of the effects of this dose on response rates. These results provide initial evidence for the possibility to use different response patterning under VI and FI schedules with equivalent reinforcement density for studying effects of drug treatment on reinforcement learning.


Handbook of Behavioral Neuroscience | 2016

Dimensions of GSK3 Monoamine-Related Intracellular Signaling in Schizophrenia

Gohar Fakhfouri; Jivan Khlghatyan; Ilya Sukhanov; Raul R. Gainetdinov; Jean-Martin Beaulieu

Abstract Since the discovery of the mechanism of action of typical antipsychotics in 1975, the dopamine theory of schizophrenia remains one of the principal doctrines to explain the pathogenesis of schizophrenia. According to this theory, increased dopaminergic neurotransmission involving D2 receptors results in dysregulation of intracellular signaling mechanisms, leading to manifestations of schizophrenia. D2 receptors signal through G protein-dependent and G protein-independent pathways, the latter involving Akt/GSK3 has proved to play a major role to the schizopheric phenotypes. GSK3 is known to play central roles in the regulation of metabolism, synaptic plasticity, and neurodevelopment. Moreover, several prominent risk factors and contributors of the disease converge on these downstream effectors of D2 receptor signaling. Finally, the medications used clinically for the management of schizophrenia affect this signaling pathway. Here, we review the signaling systems altered in schizophrenia with a focus on GSK3 networks and discuss their involvement in the pathophysiology of the disease as well as their potential for the development of diagnostic and therapeutic tools.

Collaboration


Dive into the Ilya Sukhanov's collaboration.

Top Co-Authors

Avatar

Raul R. Gainetdinov

Saint Petersburg State University

View shared research outputs
Top Co-Authors

Avatar

Stefano Espinoza

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Damiana Leo

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Emanuele

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Tatiana D. Sotnikova

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Evelina Chieregatti

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Liudmila Mus

Istituto Italiano di Tecnologia

View shared research outputs
Researchain Logo
Decentralizing Knowledge