Skip to main content
Eliyahu Dremencov
  • Institute of Molecular Physiology and Genetics
    Centre for Biosciences
    Slovak Academy of Sciences
    Dúbravská cesta 9
    840 05 Bratislava
    Slovak Republic
  • +421915033639
  • Eliyahu Dremencov, MMedSc, PhD, is a Head of Meuropharmacological Laboratory within the Institute of Molecular Physio... moreedit
  • Pierre Blier, MD, PhD, Gal Yadid, PhD, Bernard Lerer, MD, Eitan Gur, MD, Michael Newman, PhDedit
The arcuate nucleus (ARN) of the hypothalamus is involved in multiple biological functions, such as feeding, sexual activity, and the regulation of the cardiovascular system. It was reported that leptin increased c-Fos expression in the... more
The arcuate nucleus (ARN) of the hypothalamus is involved in multiple biological functions, such as feeding, sexual activity, and the regulation of the cardiovascular system. It was reported that leptin increased c-Fos expression in the proopiomelanocortin (POMC)-and decreased it in the neuropeptide-Y (NPY)-positive neurons of the ARN, suggesting that it stimulates the former, and inhibits the later. This study aimed at the direct electrophysiological examination of the effect of leptin on ARN neurons and to investigate potential sex-dimorphic changes. Wistar rats were anesthetized with urethane and the electrodes were inserted into the ARN. After a spontaneous active neuron was recorded for at least one minute, leptin was administered intravenously, and the firing activity of the same neuron was recorded for two additional minutes. It was found that approximately half of the ARN neurons had an excitatory, and another half an inhibitory response to the leptin administration. The excitability of the neurons with excitatory response to leptin was not different between the sexes. The average firing rate of the neurons with inhibitory response to leptin in females was, however, significantly lower comparing to the males. The obtained results demonstrate that the ARN neurons with stimulatory response to leptin are POMC and those with inhibitory response are NPY neurons. NPY Y1 receptor be might responsible, at least in part, for the sex differences in the excitability of the neurons putatively identified as NPY neurons.
Background: There is growing evidence that the treatment of several mental disorders can potentially benefit from activation of delta-opioid receptors. In the future, delta-agonists with a safe pharmacological profile can be used for the... more
Background: There is growing evidence that the treatment of several mental disorders can potentially benefit from activation of delta-opioid receptors. In the future, delta-agonists with a safe pharmacological profile can be used for the treatment of mood disorders in pregnant women. However, the data on prenatal exposure to delta-opioid agonists are missing. The present study is aimed to test the hypothesis that the activation of delta-opioid receptors during gravidity has positive effects on the behaviour accompanied by changes in glutamate and monoamine neurotransmission. Methods: Gestating Wistar rats were chronically treated with a selective deltaagonist SNC80 or vehicle. Adult male and female offspring underwent novel object recognition (for the assessment of cognition) and open field (for the assessment of anxiety and habituation) tests, followed by in vivo electrophysiological examination of the activity of hippocampal glutamate and midbrain serotonin (5-HT) and dopamine neurons. Results: We found that the maternal treatment with SNC80 did not affect the offspring's anxiety, habituation, and 5-HT neuronal firing activity. Female offspring of SNC80-treated dams exhibited improved novelty recognition associated with decreased firing rate and burst activity of glutamate and dopamine neurons. Conclusion: Maternal treatment with delta-opioid agonists during gestation may have a pro-cognitive effect on offspring without any negative effects on anxiety and habituation. The putative pro-cognitive effect might be mediated via mechanism(s) involving the firing activity of hippocampal glutamate and mesolimbic dopamine neurons.
Exposure by women to stressors before pregnancy increases their risk of contracting prenatal depression, a condition which typically may require antidepressant treatment. And even though such perinatal antidepressant treatment is... more
Exposure by women to stressors before pregnancy increases their risk of contracting prenatal depression, a condition which typically may require antidepressant treatment. And even though such perinatal antidepressant treatment is generally considered to be safe. For the mother, its effects on the development and functioning of the offspring`s brain remain unknown. In this study, we aimed to investigate the effects of pregestational chronic unpredictable stress (CUS) and perinatal bupropion on the anxiety behavior and firing activity of the dorsal raphe nucleus (DRN) serotonin (5-HT) neurons. Female rats underwent CUS for three weeks before mating. Bupropion was administered to them from gestation day ten until their offspring were weaned. Behavioral (elevated plus maze or EPM test) and neurophysiological (single-unit in vivo electrophysiology) assessments were performed on offspring who reached the age of 48-56 days. We found that maternal CUS and perinatal bupropion, as separate factors on their own, did not change offspring behavior. There was, however, an interaction between their effects on the number of entries to the open arms and time spent in the intersection: maternal CUS tended to decrease these values, and perinatal bupropion tended to diminish CUS effect. Maternal CUS increased the firing activity of 5-HT neurons in males, but not females. Perinatal bupropion did not alter the firing activity of 5-HT neurons but tended to potentiate the maternal CUS-induced increase in 5-HT neuronal firing activity. The CUS-induced increase in firing activity of 5-HT neurons might be a compensatory mechanism that diminishes the negative effects of maternal stress. Perinatal bupropion does not alter the offspring`s anxiety and firing activity of 5-HT, but it does intervene in the effects of maternal stress.
Fibroblast growth factor 2 (FGF2) is involved in the development and maintenance of the brain dopamine system. We previously showed that alcohol exposure alters the expression of FGF2 and its receptor, FGF receptor 1 (FGFR1) in mesolimbic... more
Fibroblast growth factor 2 (FGF2) is involved in the development and maintenance of the brain dopamine system. We previously showed that alcohol exposure alters the expression of FGF2 and its receptor, FGF receptor 1 (FGFR1) in mesolimbic and nigrostriatal brain regions, and that FGF2 is a positive regulator of alcohol drinking. Here, we determined the effects of FGF2 and of FGFR1 inhibition on alcohol consumption, seeking and relapse, using a rat operant self-administration paradigm. In addition, we characterized the effects of FGF2-FGFR1 activation and inhibition on mesolimbic and nigrostriatal dopamine neuron activation using in vivo electrophysiology. We found that recombinant FGF2 (rFGF2) increased the firing rate and burst firing activity of dopaminergic neurons in the mesolimbic and nigrostriatal systems and led to increased operant alcohol selfadministration. In contrast, the FGFR1 inhibitor PD173074 suppressed the firing rate of these dopaminergic neurons, and reduced operant alcohol self-administration. Alcohol seeking behavior was not affected by PD173074, but this FGFR1 inhibitor reduced post-abstinence relapse to alcohol consumption, albeit only in male rats. The latter was paralleled by the increased potency and efficacy of PD173074 in inhibiting dopamine neuron firing. Together, our findings suggest that targeting the FGF2-FGFR1 pathway can reduce alcohol consumption, possibly via altering mesolimbic and nigrostriatal neuronal activity.
It was previously reported that the delta opioid receptor (DOR) agonist SNC80 and antagonist naltrindole modulate the excitability of hippocampal glutamate neurons in primary cultures. The present study aimed to investigate the acute... more
It was previously reported that the delta opioid receptor (DOR) agonist SNC80 and antagonist naltrindole modulate the excitability of hippocampal glutamate neurons in primary cultures. The present study aimed to investigate the acute effects of these ligands on the firing activity of hippocampal cornu ammonis 1/3 (CA1/3) glutamate, dorsal raphe nucleus (DRN) serotonin (5-HT), locus coeruleus (LC) noradrenaline, and ventral tegmental area (VTA) dopamine neurons in in vivo conditions. Adult Wistar male rats were used. SNC80 and naltrindole were administered intravenously. Neuronal firing activity was assessed using extracellular single-unit electrophysiology. SNC80, administered first at 1-3 mg/kg, dose-dependently inhibited CA1/3 glutamate, DRN 5-HT, and VTA dopamine neurons. Naltrindole, administered at 1-3 mg/kg after SNC80, did not have any additional effect. Naltrindole, administered first at 1-3 mg/kg, stimulated DRN 5-HT neurons in a dose-dependent manner; this stimulation was dose-dependently reversed by 1-3 mg/kg of SNC80. SNC80 and naltrindole inhibited LC noradrenaline neurons when only they were co-administered at 3 mg/kg, and only when SNC80 was administered first. In conclusion, DOR ligands alter the firing activity of hippocampal glutamate and brainstem monoamine neurons in in vivo conditions. The psychoactive effects of DOR ligands, reported in previous studies, might be explained, at least in part, by their ability to modulate the firing activity of hippocampal glutamate and brainstem monoamine neurons.
Background Short-term treatment with non-peptide agonists of delta-opioid receptors, such as agonist SNC80, induced behavioral effects in rodents, which could be modulated via changes in central neurotransmission. The present experiments... more
Background Short-term treatment with non-peptide agonists of delta-opioid receptors, such as agonist SNC80, induced behavioral effects in rodents, which could be modulated via changes in central neurotransmission. The present experiments aimed at testing the hypothesis that chronic treatment with SNC80 induces anxiolytic effects associated with changes in hippocampal glutamate and brainstem monoamine pathways. Methods Adult male Wistar rats were used in experiments. Rats were treated with SNC80 (3 mg/kg/day) for fourteen days. Neuronal excitability was assessed using extracellular in vivo single-unit electrophysiology. The behavioral parameters were examined using the elevated plus maze and open field tests. Results Chronic SNC80 treatment increased the excitability of hippocampal glutamate and ventral tegmental area dopamine neurons and had no effect on the firing activity of dorsal raphe nucleus serotonin cells. Chronic SNC80 treatment induced anxiolytic effects, which were, however, confounded by increased locomotor activity clearly confirmed in an open field test. The ability to cope with stressful situations and habituation processes in a novel environment was not influenced by chronic treatment with SNC80. Conclusion Our study suggests that the psychoactive effects of SNC80 might be explained by its ability to stimulate hippocampal glutamate and mesolimbic dopamine transmission.
Perinatal depression is the most common psychiatric complication of pregnancy, with its detrimental effects on maternal and infant health commonly underrated. There is a pressing need for specific molecular biomarkers, with... more
Perinatal depression is the most common psychiatric complication of pregnancy, with its detrimental effects on maternal and infant health commonly underrated. There is a pressing need for specific molecular biomarkers, with pregnancy-related decline in brain-derived neurotrophic factor (BDNF) in the blood and downregulation of TrkB receptor in the brain reported in both clinical and preclinical studies. In this review, we highlight the emerging role of BDNF in reproductive biology and discuss evidence suggesting its deficiency as a risk factor for perinatal depression. With the increasing evidence for restoration of serum BDNF levels by antidepressant therapy, the strengthening association of perinatal depression with deficiency of BDNF advocates its potential as a surrogate endpoint for preclinical and clinical studies.
Trace amine-associated receptor 1 (TAAR1) has been recently identified as a target for the future antidepressant, antipsychotic, and anti-addiction drugs. Full (e.g. RO5256390) and partial (e.g. RO5263397) TAAR1 agonists showed... more
Trace amine-associated receptor 1 (TAAR1) has been recently identified as a target for the future antidepressant, antipsychotic, and anti-addiction drugs. Full (e.g. RO5256390) and partial (e.g. RO5263397) TAAR1 agonists showed antidepressant-, antipsychotic-and anti-addiction-like behavioral effects in rodents and primates. Acute RO5256390 suppressed, and RO5263397 stimulated serotonin (5-HT) neurons of the dorsal raphe nucleus (DRN) and dopamine neurons of the ventral tegmental area (VTA) in brain slices, suggesting that the behavioral effects of TAAR1 ligands involve 5-HT and dopamine. For more comprehensive testing of this hypothesis, we examined acute and chronic effects of RO5256390 and RO5263397 on monoamine neurons in in vivo conditions. Excitability of 5-HT neurons of the DRN, noradrenaline neurons of the locus coeruleus (LC), and dopamine neurons of the VTA was assessed using single-unit electrophysiology in anesthetized rats. For acute experiments, RO5256390 and RO5263397 were administered intravenously; neuronal excitability after RO5256390 and RO5263397 administration was compared to the basal activity of the same neuron. For chronic experiments, RO5256390 was administered orally for fourteen days prior to electrophysiological assessments. The neuronal excitability in RO5256390-treated rats was compared to vehicle-treated controls. We found that acute RO5256390 inhibited 5-HT and dopamine neurons. This effect of RO5256390 was reversed by the subsequent and prevented by the earlier administration of RO5263397. Acute RO5256390 and RO5263397 did not alter the excitability of LC noradrenaline neurons in a statistically significant way. Chronic RO5256390 increased excitability of 5-HT neurons of the DRN and dopamine neurons of the VTA. In conclusion, the putative antidepressant and antipsychotic effects of TAAR1 ligands might be mediated, at least in part, via the modulation of excitability of central 5-HT and dopamine neurons.
Background: Exposure to predator scent (PS) has been used as a model of stress associated with danger to life and body integrity. Under stress conditions, the brain serotoninergic (5-HT) system plays an important role. Methods: We tested... more
Background: Exposure to predator scent (PS) has been used as a model of stress associated with danger to life and body integrity. Under stress conditions, the brain serotoninergic (5-HT) system plays an important role. Methods: We tested the hypothesis that repeated PS exposure alters the excitability of 5-HT neurons of the dorsal raphe nucleus. To study the mechanisms involved, we approached serum and adrenal corticosterone and aldosterone concentrations, as well as brain-derived neurotrophic factor (BDNF) expression. Adult male Sprague-Dawley rats were exposed to PS for ten minutes daily for ten consecutive days. Two weeks after the last exposure, electrophysiological and biochemical assessments were performed. Results: Measurements by in vivo electrophysiology showed increased firing activity of 5-HT neurons in rats exposed to PS. Exposure to PS resulted in reduced serum corticosterone and aldosterone concentrations. Concentrations of both corticosteroids in the adrenal glands and the relative weight of the adrenals were unaffected. The gene expression of hippocampal BDNF of rats exposed to PS remained unaltered. PS exposure failed to induce changes in the gene expression of selected adrenal steroidogenic factors. Conclusion: Reduced corticosteroid concentrations in the blood appear to be the result of increased metabolism and/or tissue uptake rather than altered steroidogenesis. The decrease in circulating corticosterone in rats who experienced repeated PS may represent part of the mechanisms leading to increased excitability of 5-HT neurons. The increase in 5-HT neuronal activity might be an important compensatory mechanism designated to diminish the harmful effects of the repeated PS exposure on the brain.
The concentrations of circulating glucocorticoids are regulated by their synthesis and metabolism. Cytochrome P450 (CYP), primarily expressed in the liver, is one of the main metabolizers of glucocorticoids. Since glucocorticoids, as well... more
The concentrations of circulating glucocorticoids are regulated by their synthesis and metabolism. Cytochrome P450 (CYP), primarily expressed in the liver, is one of the main metabolizers of glucocorticoids. Since glucocorticoids, as well as monoamines, are fundamental in stress, the link between hepatic glucocorticoid metabolism and central monoamine transmission might be important in pathophysiology of stress-related disorders. We had previously reported that CYP inhibition by proadifen (SKF525) led to the inhibition of central serotonin (5-HT) neurons. The aim of this study was to investigate the effect of SKF525 on the excitability of central catecholamine neurons. Adult male Wistar rats were administered SKF525 forty-eight, twenty-four, and one hour before electrophysiological assessments. Control animals were injected saline. Rats were anesthetized with chloral hydrate and glass electrodes were inserted into the locus coeruleus (LC) or ventral tegmental area (VTA). Noradrenaline neurons of the LC and dopamine of the VTA neurons were identified, and their firing activity was recorded. It was found that the SKF525 enhanced the excitability of noradrenaline and reduced the excitability of dopamine neurons. We suggest that corticosterone-induced inhibition of 5-HT neurons underlines, at least in part, the ability of SKF525 to stimulate noradrenaline neurons. The inhibitory effect of SKF525 on dopamine neurons might be in turn secondary to the stimulatory effect of this compound on noradrenaline neurons.
Depression associated with poor general medical condition, such as post-stroke (PSD) or post-myocardial infarction (PMID) depression, is characterized by resistance to classical antidepressants. Special treatment strategies should thus be... more
Depression associated with poor general medical condition, such as post-stroke (PSD) or post-myocardial infarction (PMID) depression, is characterized by resistance to classical antidepressants. Special treatment strategies should thus be developed for these conditions. Our study aims to investigate the mechanism of action of 2-morpholino-5-phenyl-6H-1,3,4-thiadiazine, hydrobromide (L-17), a recently designed thiadiazine derivative with putative neuro- and cardioprotective and antidepressant-like effects, using combined in silico (for prediction of the molecular binding mechanisms), ex vivo (for assessment of the neural excitability using c-Fos immunocytochemistry), and in vivo (for direct examination of the neuronal excitability) methodological approaches. We found that the predicted binding affinities of L-17 to serotonin (5-HT) transporter (SERT) and 5-HT3 and 5-HT1A receptors are compatible with selective 5-HT serotonin reuptake inhibitors (SSRIs) and antagonists of 5-HT3 and 5-HT1A receptors, respectively. L-17 robustly increased c-Fos immunoreactivity in the amygdala and decreased it in the hippocampus. L-17 dose-dependently inhibited 5-HT neurons of the dorsal raphe nucleus; this inhibition was partially reversed by the 5-HT1A antagonist WAY100135. We suggest that L-17 is a potent 5-HT reuptake inhibitor and partial antagonist of 5-HT3 and 5-HT1A receptors; the effects of L-17 on amygdaloid and hippocampal excitability might be mediated via 5-HT, and putatively mediate the antidepressant-like effects of this drug. Since L-17 also possesses neuro- and cardioprotective properties, it can be beneficial in PSD and PMID. Combined in silico predictions with ex vivo neurochemical and in vivo electrophysiological assessments might be a useful strategy for early assessment of the efficacy and neural mechanism of action of novel CNS drugs.
Notwithstanding major advances in psychotherapeutics, their efficacy and specificity remain limited. The slow onset of beneficial outcomes and numerous adverse effects of widely used medications remain of chief concern, warranting... more
Notwithstanding major advances in psychotherapeutics, their efficacy and specificity remain limited. The slow onset of beneficial outcomes and numerous adverse effects of widely used medications remain of chief concern, warranting in-depth studies. The majority of frontline therapies are thought to enhance the endogenous monoaminergic drive, to initiate a cascade of events leading to lasting functional and structural plasticity. The latter also involves alterations in trophic factor signalling, including brain-derived neurotrophic factor (BDNF), NGF (nerve growth factors), vascular endothelial growth factor (VEGF), fibroblast growth factor 2 (FGF2), glial cell-derived neurotrophic factor (GDNF), and others. In several major mental disorders, emerging data suggest protective and restorative effects of trophic factors in preclinical models, when applied on their own. Antidepressant outcomes of VGF and FGF2, for instance, were shown in experimental animals, while BDNF and GDNF prove useful in the treatment of addiction, schizophrenia, and autism spectrum disorders. The main challenge with the effective translation of these and other findings in the clinic is the knowledge gap in action mechanisms with potential risks, as well as the lack of effective platforms for validation under clinical settings. Herein, we review the state-of-the-art and advances in the therapeutic use of trophic factors in several major neuropsychiatric disorders.
There is evidence that plasma cortisol concentration can be either increased or decreased in patients with depression and related anxiety and stress-related disorders; the exact pathophysiological mechanisms of this state are not almost... more
There is evidence that plasma cortisol concentration can be either increased or decreased in patients with depression and related anxiety and stress-related disorders; the exact pathophysiological mechanisms of this state are not almost clear. Several distinct theories were proposed and mechanisms, which could lead to decreased glucocorticoid signaling and/or levels, were described. However, there is a possible drawback in almost all the theories proposed: insufficient attention to the inflammatory process, which is undoubtedly present in several stress-related disorders, including post-traumatic stress disorder (PTSD). Previous studies only briefly mentioned the presence of an inflammatory reaction's signs in PTSD, without giving it due importance, although recognizing that it can affect the course of the disease. With that, the state of biochemical changes, characterized by the low glucocorticoids, glucocorticoid receptor's resistance and the signs of the persistent inflammation (with the high levels of circulating cytokines) might be observed not only in PTSD but in coronary heart diseases and sys-temic chronic inflammatory diseases (rheumatoid arthritis) as well. That is why the present review aims to depict the pathophysiological mechanisms, which lead to a decrease in glucocorticoids in PTSD due to the action of inflammatory stimuli. We described changes in the glucocorticoid system and inflam-matory reaction as parts of an integral system, where glucocorticoids and the glucocorticoid receptor reside at the apex of a regulatory network that blocks several inflammatory pathways, while decreased glucocorticoid signaling and/or level leads to unchecked inflammatory reactions to promote patholo-gies such as PTSD. LAY SUMMARY This review emphasizes the importance of inflammatory reaction in the development of puzzling conditions sometimes observed in severe diseases including post-traumatic stress disorder-the decreased levels of glucocorticoids in the blood. Following the classical concepts, one would expect an increase in glucocorticoid hormones, since they are part of the feedback mechanism in the immune system, which reduces stress and inflammation. However, low levels of glucocorticoid hormones are also observed. Thus, this review describes potential mechanisms, which can lead to the development of such a state.
KEYWORDS Lipopolysaccharide (LPS); Prenatal immune activation; Serotonin (5-HT); Noradrenaline; Dopamine; Electrophysiology in vivo. Abstract Higher risk of depression and schizophrenia in descendants of mothers experienced acute... more
KEYWORDS Lipopolysaccharide (LPS); Prenatal immune activation; Serotonin (5-HT); Noradrenaline; Dopamine; Electrophysiology in vivo. Abstract Higher risk of depression and schizophrenia in descendants of mothers experienced acute infection during the pregnancy has been reported. Since monoamines are fundamental in mentioned psychopathologies, it is possible that maternal immune activation leads to impaired functioning of serotonin (5-HT), noradrenaline, and dopamine neurons in offspring. To test this hypothesis, we examined the effect of maternal immune activation by lipopolysaccharide (LPS) in rats on the excitability of monoamine-secreting neurons in the offspring. LPS was administered during days 15-19 of the gestation in the rising doses of 20-80 μg/kg; control dams received vehicle. During days 53-63 postpartum , rats were anesthetized and electrodes were inserted into the dorsal raphe nucleus, locus coeruleus, and ventral tegmental area for in vivo excitability assessment of 5-HT, noradrenaline, and dopamine neurons. Maternal immune activation suppressed the firing rate of 5-HT neurons in both sexes and stimulated the firing rate of dopamine neurons in males. Decrease in the firing rate of 5-HT neurons was accompanied with an increase, and Please cite this article as: K. Csatlosova, E. Bogi and B. Durisova et al., Maternal immune activation in rats attenuates the excitability of monoamine-secreting neurons in adult offspring in a sex-specific way, European Neuropsychopharmacology, https://doi. JID: NEUPSY [m6+; December 16, 2020;21:18 ] increase in the firing rate of dopamine neurons with a decrease, in the density of spontaneously active cells. Maternal immune activation also decreased the variability of interspike intervals in 5-HT and dopamine neurons. It is possible that the alteration of excitability of 5-HT and dopamine neurons by maternal immune activation is involved in the psychopathologies induced by infectious disease during the pregnancy. Stimulation of dopamine excitability in males might be a compensatory mechanism secondary to the maternal immune challenge-induced suppression of 5-HT neurons.
Atypical antipsychotic drugs were introduced in the early 1990s. Unlike typical antipsychotics, which are effective only against positive symptoms of schizophrenia, atypical antipsychotics are effective against negative and cognitive... more
Atypical antipsychotic drugs were introduced in the early 1990s. Unlike typical antipsychotics, which are effective only against positive symptoms of schizophrenia, atypical antipsychotics are effective against negative and cognitive symptoms as well. Furthermore, they are effective not only in psychotic but also in affective disorders, on their own or as adjuncts to antidepressant drugs. This review presents the neural mechanisms of currently existing atypical antipsychotics and putative antipsychotics currently being investigated in preclinical and clinical studies and how these relate to their effectiveness in mood disorders such as depression, anxiety, and post-traumatic stress disorder (PTSD). Typical antipsychotics act almost exclusively on the dopamine system. Atypical drugs, however, modulate serotonin (5-HT), norepinephrine, and/or histamine neurotransmission as well. This multimodal mechanism of action putatively underlies the beneficial effect of atypical antipsychotics in mood and anxiety disorders. Interestingly, novel experimental drugs having dual antipsychotic and antidepressant therapeutic potential, such as histamine, adenosine, and trace amine-associated receptors (TAAR) ligand, are also characterized by a multimodal stimulatory effect on central 5-HT, norepinephrine, and/or histamine transmission. The multimodal stimulatory effect on central monoamine neurotransmission may be thus primarily responsible for the combined antidepressant and antipsychotic therapeutic potential of certain central nervous system (CNS) drugs.
Background: Delta-opioid receptor (DOR)-mediated modulation of hippocampal neural networks is involved in emotions, cognition, and in pathophysiology and treatment of mood disorders. In this study, we examined the effects of DOR agonist... more
Background: Delta-opioid receptor (DOR)-mediated modulation of hippocampal neural networks is involved in emotions, cognition, and in pathophysiology and treatment of mood disorders. In this study, we examined the effects of DOR agonist (SNC80) and antagonist (naltrindole) on the excitability of individual hippocampal neurons.
Methods: Primary neuronal cultures were prepared from hippocampi of newborn rats and cultivated in vitro for 8-14 days (DIV8-14). The effects of SNC80 naltrindole on evoked and spontaneous action potentials (APs) were measured at DIV8-9 and DIV13-14, respectively.
Results: SNC80 (100 µM) potentiated spontaneous AP firing and stimulated sodium current; naltrindole had opposite effects. The stimulatory effect of 100 µM of SNC80 was revoked by pre-administration of 1 µM of naltrindole. SNC80 and naltrindole induced similar inhibitory effects on the evoked AP firing and on the calcium current. Further, SNC80 inhibited both peak and sustained potassium currents. Naltrindole had no effect on potassium currents.
Conclusion: We suggest that the effects of naltrindole and high concentration of SNC80 on the sodium currents are mediated via DORs and underlying the changes in spontaneous activity. The inhibitory effects of SNC80 on calcium and potassium currents might also be DOR-dependent; these currents might mediate SNC80 effect on the evoked AP firing. The inhibitory effects of naltrindole on calcium and of low doses of SNC80 on sodium currents might be however DOR-independent. The behavioral effects of SNC80 and naltrindole, observed in previous studies, might be mediated, at least in part, via the modulatory effect of these ligands on the excitability of hippocampal neurons.
Background: Repeated exposure to predator scent stress (PSS) has been used as an animal model of complex post-traumatic stress disorder (CPTSD). The aim of the current study was to assess brain monoamines and their primary metabolites... more
Background: Repeated exposure to predator scent stress (PSS) has been used as an animal model of complex post-traumatic stress disorder (CPTSD). The aim of the current study was to assess brain monoamines and their primary metabolites concentrations in male Wistar rats (16 control, 19 exposed to chronic PSS). Methods: Rats were exposed to PSS for ten days. Fourteen days later, the rats' anxiety index (AI) was assessed with an elevated plus maze test; based on differences in AI, the rats were segregated into low-(AI ≤ 0.8, n = 9) and high-(AI > 0.8, n = 10) anxiety phenotypes. Plasma corticosterone levels were measured by radio-immunoassay. Brain monoamines and their metabolites were measured using high-performance liquid chromatography with electrochemical detector. Results: PSS exposure led to a significant increase in average rats' AI and a reduction in plasma corticosterone levels. Medullar catecholamines and hippocampal and neocortical norepinephrine levels were increased, and pontine norepinephrine and cerebellar dopamine decreased in PSS-exposed rats. Cerebellar norepinephrine levels were increased, and midbrain, hippocampal, and neocortical 5-HT and hypothalamic and hippocampal dopamine levels-decreased in high-, but not in low-anxiety rats. The decrease in hippocampal dopamine levels was accompanied by an increase of DOPAC levels, suggesting and abnormal metabolism of this transmitter. Conclusion: Reductions in 5-HT and dopamine in mid-and forebrain brain areas are associated with stress susceptibility in rodents and perhaps also with PTSD vulnerability in humans. Dopamine and 5-HT metabolism and its modulation by glucocorticoids appear to play a role in stress susceptibility and in CPTSD.
SMe1EC2M3 is a pyridoindole derivative related to the neuroleptic drug carbidine. Based on the structural similarities of SMe1EC2M3 and known serotonin (5-HT), norepinephrine, and dopamine reuptake inhibitors, we hypothesized that this... more
SMe1EC2M3 is a pyridoindole derivative related to the neuroleptic drug carbidine. Based on the structural similarities of SMe1EC2M3 and known serotonin (5-HT), norepinephrine, and dopamine reuptake inhibitors, we hypothesized that this compound may also have triple reuptake inhibition efficacy and an antidepressant-like effect. PreADMET and Dragon software was used for in silico prediction of pharmacokinetics and pharmacodynamics of SMe1EC2M3. Forced swim test was used to evaluate its antidepressant-like effects. Extracellular in vivo electrophysiology was used to assess 5-HT, norepinephrine, and dopamine reuptake inhibition efficacy of SMe1EC2M3. PreADMET predicted reasonable intestinal absorption, plasma protein binding, and blood-brain permeability for SMe1EC2M3. Dragon forecasted its efficiency as an antidepressant. Using behavioral measurements, it was found that SMe1EC2M3 decreased immobility time and increase swimming time during the forced swim test (FST). Electrophysiological investigations showed that SMe1EC2M3 dose-dependently suppressed the excitability of 5-HT neurons of the dorsal raphe nucleus (DRN), norepinephrine neurons of the locus coeruleus (LC), and dopamine neurons of the ventral tegmental area (VTA). The SMe1EC2M3-induced suppression of 5-HT, norepinephrine, and dopamine neurons was reversed by the antagonists of serotonin-1A (5-HT1A; WAY100135), α-2 adrenergic (α2, yohimbine), and dopamine-2 receptors (D2, haloperidol), respectively. We conclude that SMe1EC2M3 is prospective triple 5-HT, norepinephrine, and dopamine reuptake inhibitor with antidepressant-like properties, however future studies should be performed to complete the pharmacological profiling of this compound.
Post-traumatic stress disorder (PTSD) is a severe psychiatric illness which may develop in individuals exposed to trauma [1], including exposure to military combat, interpersonal violence, and childhood maltreatment. The disorder is... more
Post-traumatic stress disorder (PTSD) is a severe psychiatric illness which may develop in individuals exposed to trauma [1], including exposure to military combat, interpersonal violence, and childhood maltreatment. The disorder is characterized by an alternating pattern of re-experiencing symptoms, avoidance of traumatic stimuli, changes in mood and cognition, numbing of general responsiveness, and increased arousal (DSM 5).

Knowledge of the PTSD biology continues to expand whereby earlier observations of elevated catecholamines and decreased cortisol in urine of chronic PTSD of combat veterans [2] were criticized for reflecting only downstream or peripheral components of a very complex pathology [3]. Nonetheless, more recent and sophisticated studies have, by in large, supported these early observations and have provided important additional data relevant to the pathomechanism of PTSD [4]. For example, neuroimaging studies identified important neural circuits in the limbic system, including amygdala, hippocampus, and prefrontal cortex (PFC) which are involved in PTSD [5] [6] [7], but did not explain the initial neuroendocrine findings, especially of decreased urinary cortisol. The concept of allostatic load is a useful paradigm to connect PTSD-related neuroendocrine findings to altered neural circuits [8] [9], however, to date, the relationship between glucocorticoid and catecholamine metabolism in PTSD has not been studied, although monoamine oxidase (MAO)-A, the key enzyme of catecholamine metabolism is glucocorticoid-dependent [10]. Here we present a new hypothesis to explain the link between activation of glucocorticoid metabolism and depression of catecholamine metabolism among different allostatic states in PTSD.
Chronic stress during pregnancy or even prior to gestation can negatively affect offspring´s neurobehavioural development. Several studies have shown, that offspring who had experienced excessive stress during gestation had higher rates... more
Chronic stress during pregnancy or even prior to gestation can negatively affect offspring´s neurobehavioural development. Several studies have shown, that offspring who had experienced excessive stress during gestation had higher rates of cognitive and mood disorders later during adolescence or in adulthood. Hippocampal neu-rons play a crucial role in the regulation of behavior, mainly in anxiety-related behaviors and spatial learning and memory. Recently, it has been shown, that excessive stress even prior to gestation could interfere with sensitive developmental processes in the brain and may affect hippocampal functioning with severe neurobe-havioural consequences in later life. The aim of this work was to investigate the effects of pre-gestational stress of the rat dams on the hippocampal excitability of the pups right after the birth. Neurobehavioural consequences of pre-gestational stress were analyzed during adolescence (35-40 postnatal days) and in early adulthood (75-80 postnatal days). We have shown that even pre-gestational chronic maternal stress increased resting membrane potential, suppressed depolarization-activated action potential firing, and increased spontaneous activity of hippocampal cells from newborn offspring. Altered function of hippocampus was reflected at the behavioural level. Adolescent male offspring of dams exposed stress prior to conception showed hyperactivity-like behaviour in a new stressful environment and increased anxiety-like behaviour during adulthood compared to adult males from non-stress group. Together, this work suggests, that chronic stress even prior to gestation can interfere with functional brain development of the offspring and can cause long-term behavioural changes at the level of neurobehavioural adaptations.
Glucocorticoid signaling is fundamental in healthy stress coping and in the pathophysiology of stress-related diseases, such as post-traumatic stress disorder (PTSD). Glucocorticoids are metabolized by cytochrome P450 (CYP) as well as... more
Glucocorticoid signaling is fundamental in healthy stress coping and in the pathophysiology of stress-related diseases, such as post-traumatic stress disorder (PTSD). Glucocorticoids are metabolized by cytochrome P450 (CYP) as well as 11-β-hydroxysteroid dehydrogenase type 1 (11βHSD1) and 2 (11βHSD2). Acute stress-induced increase in glucocorticoid concentrations stimulates the expression of several CYP sub-types. CYP is primarily responsible for glucocorticoid metabolism and its increased activity can result in decreased circulating glucocorticoids in response to repeated stress stimuli. In addition, repeated stress-induced glucocorticoid release can promote 11βHSD1 activation and 11βHSD2 inhibition, and the 11βHSD2 suppression can lead to apparent mineralocorticoid excess. The activation of CYP and 11βHSD1 and the suppression of 11βHSD2 may at least partly contribute to development of the blunted glucocorticoid response to stressors characteristic in high trait anxiety, PTSD and other stress-related disorders. Glucocorticoids and glucocorticoid-metabolizing enzymes interact closely with other biomolecules such as inflammatory cytokines, monoamines and some monoamine-metabolizing enzymes, namely the monoamine oxidase type A (MAO-A) and B (MAO-B). Glucocorticoids boost MAO activity and this decreases monoamine levels and induces oxidative tissue damage which then activates inflammatory cytokines. The inflammatory cytokines suppress CYP expression and activity. This dynamic cross-talk between glucocorticoids, monoamines and their metabolizing enzymes could be a critical factor in the pathophysiology of stress-related disorders.
The concepts of allostatic load and overload, i.e. a dramatic increase in the allostatic load that predisposes to disease, have been extensively described in the literature. Here, we show that rats engaging in active offensive response... more
The concepts of allostatic load and overload, i.e. a dramatic increase in the allostatic load that predisposes to disease, have been
extensively described in the literature. Here, we show that rats engaging in active offensive response (AOR) behavioral strategies
to chronic predator scent stress (PSS) display less anxiety behavior and lower plasma cortisol levels versus rats engaging in
passive defensive response (PDR) behavioral strategies to chronic PSS. In the same chronic PSS paradigm, AOR rats also have higher
lactate and lower glutamate levels in amygdala but not in control-region hippocampus versus PDR rats. The implications of these
findings for regulation of allostatic and stress responses, and post-traumatic stress disorder (PTSD) are discussed.
The aim of this study was to investigate the effect of chronic predator scent stress (PSS) on monoamine levels in rat thalamus and hypothalamus. Rats were exposed to the PSS (sand containing cat urine) for ten minutes daily for ten days.... more
The aim of this study was to investigate the effect of chronic predator scent stress (PSS) on monoamine levels in rat thalamus and hypothalamus. Rats were exposed to the PSS (sand containing cat urine) for ten minutes daily for ten days. Control animals were exposed to the sand containing clean water. Fifteen days later, rats’ behavior and thalamic and hypothalamic levels of monoamines were analyzed. PSS rats had elevated anxiety, increased thalamic serotonin and decreased hypothalamic dopamine concentrations. This decrease in hypothalamic dopamine may explain, at least in part, lowered corticosterone levels observed in PSS animals in our previous studies.
The aim of this study was to investigate the effect an inhibitor of cytochrome-P450, proadifen hydrochloride (SKF525), on the excitability of serotonin neurons. Adult male Wistar rats were administered SKF525 forty-eight, twenty-four, and... more
The aim of this study was to investigate the effect an inhibitor of cytochrome-P450, proadifen hydrochloride (SKF525), on the excitability of serotonin neurons. Adult male Wistar rats were administered SKF525 forty-eight, twenty-four, and one hour before electrophysiological assessments.  Control animals were injected saline.  Rats were anesthetized with chloral hydrate and glass electrodes were stereotaxically inserted into the dorsal raphe nucleus (DRN). Serotonin neurons were identified and their firing activity was recorded.  It was found that the SKF525 inhibits the excitability of 5-HT neurons. We suggest that corticosterone might play a key role in the SKF525-induced inhibition of 5-HT neurons.
Depression is a severe disorder of the central nervous system (CNS), characterized by multiple emotional, cognitive, and neuroendocrine malfunctions. The lack of the objective diagnostic means for depression and limited efficacy of the... more
Depression is a severe disorder of the central nervous system (CNS), characterized by multiple emotional, cognitive, and neuroendocrine malfunctions. The lack of the objective diagnostic means for depression and limited efficacy of the existing antidepressants make depression the second major reason for disability and the costliest medical condition in Europe. There is a strong evidence that depression is an organic disease of the CNS rather than simple mood perturbation. However, the exact biomarkers directly linked with depression, such as Lewy Bodies in Parkinson disease, not yet discovered. It can be hypothesizing that the organic brain abnormalities directly linked with depression can be detected on the level of subcellular neuronal structures, such as dendritic spines. Four lines of evidence support the hypothesis that dendritic spines are involved in pathophysiology of depression. Firstly, dendritic spines are the basic information processing units of the brain, and depression can be seen a disorder characterized by impaired information processing. Secondly, dendritic spines are heavily expressed in the limbic areas of the brain, which are responsible for the brain functions impaired in depression. Thirdly, dendritic spines are regulated by monoamines, which are primary target of all known antidepressant drugs. Finally, there are initial reports that antidepressant drugs amend density and morphology of dendritic spines in animal models.
It was previously reported that adenosine-2A (A2A) receptors interact with dopamine-2 (D2) receptors on a molecular level. The aim of the current study was to investigate the functional output of this interaction. In vivo microdialysis... more
It was previously reported that adenosine-2A (A2A) receptors interact with dopamine-2 (D2) receptors on a molecular level. The aim of the current study was to investigate the functional output of this interaction. In vivo microdialysis was used to assess the effects of an antagonist of A 2A receptors, ZM 241385, and an antagonist of D2 receptors haloperidol, either alone or in combination, on brain catecholamine levels. It was found that ZM 241385 did not alter catecholamine levels by its own, but potentiated haloperidol-induced dopamine and norepinephrine release in the nucleus accumbens and prefrontal cortex, respectively. In vivo electrophysiology was used to assess the effect of an agonist (CGS 216820) and an antagonist (ZM 241385) of A 2A receptors on the excitability of dopamine and norepinephrine neurons. It was found that CGS 216820 dose-dependently inhibited dopamine and norepinephrine neurons and ZM 241385 reversed this inhibition. In conclusion, those A 2A receptors modulate brain catecholamine transmission, and this modulation is mediated, at least in part, via the regulation of excitability of norepinephrine and dopamine neurons. The ability of antagonists of A2A receptors to potentiate the effect of haloperidol on brain norepinephrine and dopamine levels may enhance its clinical efficacy as an antipsychotic drug.
Background: The antidepressant effect of physical exercise has been reported in several clinical and animal studies. Since serotonin (5-HT), norepinephrine and dopamine play a central role in depression, it is possible that the beneficial... more
Background: The antidepressant effect of physical exercise has been reported in several clinical and animal studies. Since serotonin (5-HT), norepinephrine and dopamine play a central role in depression, it is possible that the beneficial effects of physical exercise are mediated via monoamine pathways. This study investigates the effects of voluntary wheel running (VWR) on the excitability of monoamine neurons.
Material and methods: Male Sprague-Dawley rats were used in the study. Voluntary wheel running (VWR) rats were housed in individual cages with free access to a running wheel, while control animals were housed in standard laboratory cages. After three weeks, the rats were anesthetized and in vivo electrophysiological recordings
were taken from dorsal raphe nucleus 5-HT neurons, locus coeruleus norepinephrine neurons and ventral tegmental dopamine neurons.
Results: VWR stimulated activity in 5-HT, but not in norepinephrine or dopamine neurons. Subsequently, acute administration of the selective serotonin reuptake inhibitor (SSRI) escitalopram in control rats led to complete suppression of 5-HT neurons; this suppression was reversed by subsequent administration of selective antagonist of 5-HT1A receptors, WAY100135. Escitalopram induced only partial inhibition of 5-HT neurons in the VWR rats while WAY100135 increased the firing
activity of 5-HT neurons above the baseline value.
Conclusions: The beneficial effect of physical exercise on mood is mediated, at least in part, via activation of 5-HT neurons. Physical exercise can potentiate the response to SSRIs by increasing the basal firing activity and by diminishing SSRI induced inhibition of 5-HT neurons.
Aim. The current work aims to summarize the experimental data (from author’s and other laboratories) on the neuronal mechanism of the mood effect of physical exercise and to suggest a potential mood-stabilizing mechanism of physical... more
Aim. The current work aims to summarize the experimental data (from author’s and other laboratories) on the neuronal mechanism of the mood effect of physical exercise and to suggest a potential mood-stabilizing mechanism of physical activity. Materials and Methods. Physical Depression is a brain disorder characterized by severe emotional, cognitive, neuroendocrine, and somatic dysfunctions. Based on this concept, the author performed a review of experimental data on various
ways to manage depression including medical treatment and physical exercise. Results. Although the last generation of antidepressant drugs demonstrates improved clinical efficacy and safety, the onset of their clinical effect is still significantly delayed from the beginning of the treatment course, and significant number of patients show lack of adequate response to these drugs and/or relapse of the disease even after initially successful treatment. Certain non-pharmaceutical strategies are used as adjuncts or replacements to antidepressant drugs when the formers are ineffective. One of such strategies is the voluntary physical exercise. Conclusions. Voluntary physical exercise can
be an adjunct physiotherapeutic treatment in depression, given together with the pharmacotherapy, e.g., with SSRIs. One of the potential mechanisms of action of physical exercise in depression is stimulation of basic and/or reversal of the SSRI-induced inhibition of 5-HT tone. Other potential mechanisms, such as neuropeptide pathways, should be investigated in the future studies.
Aim of the review is to summarize the contemporary evidences on interactions between brain neurotransmitters and their role in pathophysiology and treatment of depression. Introduction. Brain neurotransmitters are biological molecules... more
Aim of the review is to summarize the contemporary evidences on interactions between brain neurotransmitters and their role in pathophysiology and treatment of depression.

Introduction. Brain neurotransmitters are biological molecules responsible for signal transduction between the neurons. Mammalian brain neurotransmitters belong to the different types of biological molecules, such as amino acids (glutamate and γ-aminobutyric acid, or GABA), monoamines (serotonin: 5-HT, norepinephrine: NE, dopamine: DA, and histamine: HA),
neuropeptides (β-endorphin, neurokinin, vasopressin, and oxytocin), or nucleotides (adenosine).

Materials and Methods. The author analyzes research papers dedicated to neurotransmitters
and their functions in mood regulation and published between 1953 and 2014. The paper focuses on evaluation of data on mechanisms of interactions between different neurotransmitters and their role in development and treatment of certain mental disorders.

Results. It has been reported that different neurotransmitters, often belonging to the different
types of biological molecules, interact on behavioral, functional, system and molecular levels. These interactions play an important role in pathophysiology of brain diseases, particularly, depression and stress and anxiety-related disorders. Conclusion. Literally all existing antidepressant drugs act on monoamine systems of the brain (5-HT, NE, and DA). Although the last-generation antidepressants and mood stabilizers demonstrated higher safety and efficacy, their therapeutic potential remains limited. The brain adenosine neurotransmission is also a potential target for the future antidepressant, mood stabilizing and antipsychotic drugs. However, these drugs may have severe side effects, for example, on cardiac activity.
It is necessary to perform further research and clinical trials to find a solution to the existing
difficulties.
Research Interests:
Depression is a brain disorder characterized by severe emotional, cognitive, neuroendocrine, and somatic dysfunctions. Although the last generation of antidepressant drugs demonstrate improved clinical efficacy and safety, the onset of... more
Depression is a brain disorder characterized by severe emotional, cognitive, neuroendocrine, and somatic dysfunctions. Although the last generation of antidepressant drugs demonstrate improved clinical efficacy and safety, the onset of their clinical effect is still significantly delayed from the beginning of the treatment course, and significant number of patients show lack of adequate response to these drugs and/or relapse of the disease even after initially successful treatment. Therefore, there is a need to develop new antidepressant drugs and/or adjuncts to existing ones. Recent studies suggest that the beneficial effect of antidepressant drugs is mediated, at least in part, via the stimulation of adult hippocampal neurogenesis and subsequent increase in hippocampal plasticity. The stimulatory effect of antidepressant drugs on hippocampal neurogenesis involves G-protein coupled receptors (GPCR) and voltage-dependent calcium channels (VDCC). It is therefore possible that the future antidepressant drugs will directly target specific GPCR and VDCC. Potential advantages and limitations of these treatment strategies are discussed in the article.
Brain monoamines (serotonin, norepinephrine, dopamine, and histamine) play an important role in emotions, cognition and in pathophysiology and treatment of mental disorders. The interactions between serotonin, norepinephrine and dopamine... more
Brain monoamines (serotonin, norepinephrine, dopamine, and histamine) play an important role in emotions, cognition and in pathophysiology and treatment of mental disorders. The interactions between serotonin, norepinephrine and dopamine were studied in numerous works, however, histamine system received less attention. The aim of this study was to investigate the interactions between histamine and other monoamines, using in vivo microdialys and electrophysiology. It was found that the inverse agonist of histamine-3 receptors, thioperamide, increased the firing activity of dopamine neurons in the ventral tegmental area. Selective agonist of histamine-3 receptors, immepip, reversed thiperamide-induced stimulation of firing activity of dopamine neurons. The firing rates of serotonin and norepinephrine neurons were not attenuated by immepip or thioperamide. Thioperamide robustly and significantly increased extracellular concentrations of serotonin, norepinephrine, and dopamine in the rat prefrontal cortex and slightly increased norepinephrine and dopamine levels in the tuberomammillary nucleus of the hypothalamus. It can be concluded that histamine stimulates serotonin, norepinephrine, and dopamine transmission in the brain. Modulation of firing of dopamine neurons is a key element in functional interactions between histamine and other monoamines. Antagonists of histamine-3 receptors, because of their potential ability to stimulate monoamine neurotransmission, might be beneficial in the treatment of mental disorders.
Many patients with major depression do not respond to selective serotonin reuptake inhibitors (SSRIs). Lack of response could be due to inhibition of dopamine (DA) release by SSRIs through 5-HT2C receptors. Combining an SSRI with a 5-HT2C... more
Many patients with major depression do not respond to selective serotonin reuptake inhibitors (SSRIs). Lack of response could be due to inhibition of dopamine (DA) release by SSRIs through 5-HT2C receptors. Combining an SSRI with a 5-HT2C antagonist may result in improved efficacy by causing simultaneous increases of serotonin (5-HT) and DA. In order to test this augmentation strategy, male Wistar rats were treated (s.c.) with an acute dose of the SSRI citalopram (Cit, 5 mg/kg), the 5-HT2C antagonist SB 242084 (SB, 2 mg/kg), or Cit + SB, and the effects on 5-HT and DA release in the nucleus accumbens (NAcc) was assessed by microdialysis. In a separate experiment, animals were treated with vehicle, Cit (20 mg/kg/d), SB (2 mg/kg/d) or Cit + SB for a period of 2 days (s.c.), and the impact on the release of 5-HT and DA in the ventral tegmental area (VTA) and NAcc was studied.. On the day of microdialysis, 5-HT2C receptor sensitivity was assessed with an SB challenge. Acutely administered Cit + SB increased 5-HT release in the NAcc more than Cit alone. SB alone increased DA release in the NAcc (not in the VTA), but when administered together with Cit, this effect was abolished. A 2-day treatment with Cit or Cit + SB increased 5-HT release in both VTA and NAcc. Combining Cit with SB augmented the effect of Cit in the VTA. DA release in VTA and NAcc was only significantly increased after 2-days of treatment with Cit + SB. In conclusion, Cit + SB had synergistic effects on 5-HT and DA release after 2-days of treatment, probably related to a decreased tonic inhibition of DA release via 5-HT2C receptors. Regional differences occur and future studies should elucidate if this augmentation strategy is beneficial on the behavioural level.
Wistar-Kyoto (WKY) rats are sensitive to chronic stressors and exhibit depression-like behavior. Dorsal raphe nucleus (DRN) serotonin (5-HT) neurons projecting to the prefrontal cortex (PFC) comprise the important neurocircuitry... more
Wistar-Kyoto (WKY) rats are sensitive to chronic stressors and exhibit depression-like behavior. Dorsal raphe nucleus (DRN) serotonin (5-HT) neurons projecting to the prefrontal cortex (PFC) comprise the important neurocircuitry underlying the pathophysiology of depression. To evaluate the DRN-PFC 5-HT system in WKY rats, we examined the effects of escitalopram (ESCIT) on the extracellular 5-HT level in comparison with Wistar rats using dual-probe microdialysis. The basal levels of 5-HT in the DRN, but not in the PFC, in WKY rats was reduced as low as 30% of Wistar rats. Responses of 5-HT in the DRN and PFC to ESCIT administered systemically and locally were attenuated in WKY rats. Feedback inhibition of DRN 5-HT release induced by ESCIT into the PFC was also attenuated in WKY rats. Chronic ESCIT induced upregulation of the DRN-PFC 5-HT system in WKY rats, with increases in basal 5-HT in the DRN, responsiveness to ESCIT in the DRN and PFC, and feedback inhibition, whereas downregulation of these effects was induced in Wistar rats. Thus, the WKY rat is an animal model of depression with low activity of the DRN-PFC 5HT system. The finding that chronic ESCIT upregulates the 5-HT system in hyposerotonergic WKY rats may contribute to improved understanding of mechanisms of action of antidepressants, especially in depression with 5-HT deficiency.
Given that the Wistar Kyoto (WKY) rat is a putative animal model of depression, the present study examined the effects of acute escitaloprarn (ESCIT) on the dorsal raphe nucleus (DRN) serotonin (5-HT) neuron and behavior in WKY rats in... more
Given that the Wistar Kyoto (WKY) rat is a putative animal model of depression, the present study examined the effects of acute escitaloprarn (ESCIT) on the dorsal raphe nucleus (DRN) serotonin (5-HT) neuron and behavior in WKY rats in comparison to Wistar rats. Furthermore, the effect of the re­peated ESCIT (5 mg/kg/day s.c. for 14 days) treatment in WKY rats was assessed.
The current study aimed to investigate the effect of histamine-3 (H(3)) receptors, expressed in the tuberomammillary nucleus (TMN) of the hypothalamus and in the prefrontal cortex (PFC), on histamine neurotransmission in the rat brain.... more
The current study aimed to investigate the effect of histamine-3 (H(3)) receptors, expressed in the tuberomammillary nucleus (TMN) of the hypothalamus and in the prefrontal cortex (PFC), on histamine neurotransmission in the rat brain. The firing activity of histamine neurons in the TMN was measured using in vivo extracellular single-unit electrophysiology, under propofol anesthesia. Extracellular histamine levels were determined using the dual (PFC and TMN) probe microdialysis, in freely-moving animals. Histamine levels in dialysates were determined using high-performance liquid chromatography (HPLC) and fluorescence detection. It was found that systemic administration of the selective H(3)-agonist, immepip, decreases, and the reverse H(3) /H(4)-agonist, thioperamide, increases the firing activity of histamine neurons in the TMN and the release of histamine in TMN and PFC. Local perfusion of immepip into the TMN increased, and thioperamide decreased, histamine levels in the TMN but not in the PFC. Local perfusion of immepip into the PFC, however, decreased extracellular histamine levels in both TMN and PFC. It can be concluded that brain H(3) receptors, and especially those expressed in the PFC, play an important role in the autoregulation of histamine neurotransmission. It is possible that H(3) receptors in the PFC are expressed on pyramidal neurons projecting to the TMN, and activation of these receptors diminishes glutamate excitatory input from PFC to the TMN. As the brain histamine system has a role in pathophysiology of psychotic, affective, cognitive, sleep and eating disorders, H(3) receptors are potential targets for future CNS medications.
The present study conducted the behavioral and neurochemical comparisons of the dorsal ,raphe nucleus (DRN) serotonin (5-HT) neuron between Wistar and Wistar­ Kyoto (WKY) rat strains, to determine whether WKY can be characterized in an... more
The present study conducted the behavioral and neurochemical comparisons of the dorsal ,raphe nucleus (DRN) serotonin (5-HT) neuron between Wistar and Wistar­ Kyoto (WKY) rat strains, to determine whether WKY can be characterized in an animal model for a certain type of depres­sion as previously suggested (De La Garza and Mahoney, 2004). Furthermore, the effects of the repeated administra­tion of escitalopram (ESCIT, 5 mg/kg/day s.c. for 14 days) on the depression-like behavior and on the 5-HT neurotransmis­sion in WKY were assessed.
The dopamine receptor family consists of D1-D5 receptors (D1R-D5R), and we explored the contributions of each dopamine receptor subtype in the piriform cortex (PirC) to social interaction impairment (SII). Rats received behavioral tests... more
The dopamine receptor family consists of D1-D5 receptors (D1R-D5R), and we explored the contributions of each dopamine receptor subtype in the piriform cortex (PirC) to social interaction impairment (SII). Rats received behavioral tests or electrophysiological recording of PirC neuronal activity after injection of the D1R/D5R agonist SKF38393, the D2R/D3R/D4R agonist quinpirole, or both, with or without pretreatment with dopamine receptor antagonists, D1R or D5R antisense oligonucleotides, the cannabinoid CB1 receptor antagonist AM281, or the endocannabinoid transporter inhibitor VDM11. Systemic injection of SKF38393 and quinpirole together, but not each one alone, induced SII and increased PirC firing rate, which were blocked by D1R or D2R antagonist. Intra-PirC microinfusion of SKF38393 and quinpirole together, but not each one alone, also induced SII, which was blocked by D1R antisense oligonucleotides or D2R antagonist but not by D3R or D4R antagonist or D5R antisense oligonucleotides. SII induced by intra-PirC SKF38393/quinpirole was blocked by AM281 and enhanced by VDM11, whereas neither AM281 nor VDM11 alone affected social interaction behavior. Coadministration of SKF38393 and quinpirole produced anxiolytic effects without significant effects on locomotor activity, olfaction, and acquisition of olfactory short-term memory. These findings suggest that SII induced by coactivation of PirC D1R and D2R requires the endocannabinoid system.
Tramadol is an atypical opioid with monoamine re-uptake inhibition properties. The aim of the current study was to compare, using in vivo microdialysis, the effect of tramadol on extracellular serotonin (5-HT) and noradrenaline (NA)... more
Tramadol is an atypical opioid with monoamine re-uptake inhibition properties. The aim of the current study was to compare, using in vivo microdialysis, the effect of tramadol on extracellular serotonin (5-HT) and noradrenaline (NA) levels in the rat ventral hippocampus with the effects of the dual 5-HT/NA inhibitors (SNRIs) duloxetine and venlafaxine, the tricyclic antidepressant clomipramine, the selective 5-HT re-uptake inhibitor (SSRI) citalopram, and the selective NA re-uptake inhibitor (NRI) reboxetine. It was found that tramadol, duloxetine and venlafaxine increased extracellular levels of both, 5-HT and NA, in a dose-dependent manner. Clomipramine also increased extracellular 5-HT and NA levels, however not dose-dependently in the tested dose range. Citalopram selectively increased extracellular 5-HT levels. Reboxetine increased extracellular NA levels and also to a minimal degree 5-HT levels. It can be concluded that, albeit less efficacious, the effects of tramadol on serotonergic and noradrenergic neurotransmission resemble those of the dual 5-HT and NA re-uptake inhibitors duloxetine, venlafaxine, and clomipramine, and are different from those of the SSRI citalopram and the NRI reboxetine.
Selective serotonin (5-HT) reuptake inhibitors (SSRIs) are used as a first-line treatment in depression. However, many depressed patients are also treated with benzodiazepines to alleviate increased anxiety and sleep disturbances normally... more
Selective serotonin (5-HT) reuptake inhibitors (SSRIs) are used as a first-line treatment in depression. However, many depressed patients are also treated with benzodiazepines to alleviate increased anxiety and sleep disturbances normally associated with depression. Since benzodiazepines inhibit 5-HT neuronal firing activity, they might attenuate SSRI-induced increase in extracellular 5-HT levels. This study aimed to assess, using in-vivo microdialysis, the effects of the benzodiazepines oxazepam or temazepan on the SSRI paroxetine-induced 5-HT increase in the hippocampus of freely moving guinea-pigs. It was found that the acute systemic administration of paroxetine increased extracellular 5-HT levels. Pre-administration of oxazepam or temazepam significantly diminished the paroxetine-induced elevation of extracellular 5-HT levels (from 350% to 200% of baseline). It was concluded that benzodiazepines attenuate the ability of SSRIs to elevate hippocampal 5-HT levels. Thus, co-administration of benzodiazepines might affect the therapeutic efficacy of SSRI treatment.
There are numerous lines of evidence pointing to norepinephrine being of crucial importance in pathophysiology of anxiety and mood disorders. First, norepinephrine projections innervate the limbic system, suggesting the involvement of... more
There are numerous lines of evidence pointing to norepinephrine being of crucial importance in pathophysiology of anxiety and mood disorders. First, norepinephrine projections innervate the limbic system, suggesting the involvement of norepinephrine in the regulation of emotions and cognition. Second, norepinephrine closely interacts with serotonin and dopamine systems, which also play very important roles in the regulation of mood. Third, it has been shown that various agents which increase norepinephrine availability, such as norepinephrine reuptake inhibitors, are also effective antidepressant drugs. And fourth, the depletion of norepinephrine causes a resurgence of depressive symptoms after successful treatment with antidepressant drugs. These observations suggest that the intensification of norepinephrine transmission can be beneficial in the treatment of affective disorders. However, various psychotropic medications have indirect effect on norepinephrine transmission. This review examines the effects of psychiatric medications on the norepinephrine system and proposes how they might be used to improve treatment outcome.
Ventral tegmental area (VTA) neuronal activity plays an important role in reward-related learning and motivation. Tracing the bursting signal is important for understanding neural state and understanding communication between individual... more
Ventral tegmental area (VTA) neuronal activity plays an important role in reward-related learning and motivation. Tracing the bursting signal is important for understanding neural state and understanding communication between individual neurons. The dopaminergic system, which projects from the VTA to other regions in the mesolimbic system, is involved in hedonia and motivation. However, the role of this system in the pathophysiology of depression and its manipulation for treatment of depression has received little attention. Inter-spike interval time series were recorded from the VTA of control Sprague–Dawley and Flinders sensitive line (FSL) rats with or without 14 days of desipramine (5 mg/kg) treatment. Comparison of the firing modes of control and desipramine-treated FSL rats reveals dissimilar patterns. Desipramine treatment normalized depressive-like behavior and elevated the dopaminergic mesolimbic activity, although not to control levels. Mesolimbic neuronal activity is known to occur either in burst or in single-spike firing mode. Herein, we suggest a third mode that is characterized as a “cluster” formed from burst and post-burst activity. A significant reduction in the activity of both bursts and cluster was detected in FSL rats, which was restored by desipramine treatment.
Background: Atypical antipsychotic drugs have been used in depressed patients not responding adequately to the selective serotonin reuptake inhibitors (SSRIs). The aim of the current study was to investigate putative mechanisms of the... more
Background: Atypical antipsychotic drugs have been used in depressed patients not responding adequately to the selective serotonin reuptake inhibitors (SSRIs). The aim of the current study was to investigate putative mechanisms of the beneficial effect of atypical antipsychotic drugs during their co-administration with SSRIs. In previous electrophysiological studies, it was found that SSRIs decrease, while atypical antipsychotics increase, norepinephrine neuronal firing. Thus, the resistance to SSRIs could be explained, at least in part, by the SSRI-induced decrease of norepinephrine neuronal firing activity, and the beneficial effect of atypical antipsychotic drugs could be explained by the reversal of the above-mentioned suppression of firing.
Methods: Rats were administered the SSRI escitalopram and the atypical antipsychotic drug risperidone. Norepinephrine neuronal activity was determined using in vivo electrophysiology.
Results: Subacute and long-term escitalopram decreased, while risperidone co administered with escitalopram increased, norepinephrine neuronal firing. Attempts at reversing the escitalopram-induced decrease of firing with various selective antagonists revealed that the serotonin-2A receptor antagonistic property of risperidone may mediate the pronoradrenergic action of atypical antipsychotics in the
presence of serotonin reuptake inhibition.
Conclusions: Risperidone reverses escitalopram-induced inhibition of norepinephrine neuronal activity by a mechanism involving serotonin-
2A receptors. This reversal may explain the beneficial effect of atypical antipsychotics in treatment-resistant depression.
Dopaminergic mesolimbic and mesocortical systems are involved in hedonia and motivation, two core symptoms of depression. However, their role in the pathophysiology of depression and their manipulation to treat depression has received... more
Dopaminergic mesolimbic and mesocortical systems are involved in hedonia and motivation, two core symptoms of depression. However, their role in the pathophysiology of depression and their manipulation to treat depression has received little attention. Previously, we showed decreased limbic dopamine (DA) neurotransmission in an animal model of depression, Flinder sensitive line (FSL) rats. Here we describe a high correlation between phase-space algorithm of bursting-like activity of DA cells in the ventral tegmental area (VTA) and efficiency of DA release in the accumbens. This bursting-like activity of VTA DA cells of FSL rats is characterized by a low dimension complexity. Treatment with the antidepressant desipramine affected both the dimension complexity of cell firing in the VTA and rate of DA release in the accumbens, as well as alleviating depressive-like behavior. Our findings indicate the potential usefulness of monitoring limbic dopaminergic dynamics in combination with non-linear analysis. Decoding the functionality of the dopaminergic system may help in development of future antidepressant drugs.

And 19 more

This monograph is a collection of selected articles on the subject of mood disorders such as depression and schizophrenia. It is divided into 3 sections: 1) Research methods in psychopharmacology - which highlights some established... more
This monograph is a collection of selected articles on the subject of mood disorders such as depression and schizophrenia. It is divided into 3 sections: 1) Research methods in psychopharmacology - which highlights some established experimental techniques to study mood disorders in human and relevant animal models, 2) Pathophysiology of mood disorders – which explains the physiological and pharmacological mechanisms responsible for mood disorders and 3) New strategies for the treatment of mood disorders – a concluding section that provides recent examples on the beneficial effects of pharmacological and non-pharmacological interventions in the relief of mood disorders. The e-book serves as a primer for graduate students and researchers interested in the physiology and treatment of affective psychological disorders. Free sample is available for downloading; order the whole book at: http://benthamscience.com/ebooks/9781608054671/index.htm
All existing antidepressant drugs are characterized by relatively long delay (two-eight weeks) between the beginning of the medication and onset of therapeutic effect of the treatment. One of the goals of psycho-pharmacological research... more
All existing antidepressant drugs are characterized by relatively long delay (two-eight weeks) between the beginning of the medication and onset of therapeutic effect of the treatment. One of the goals of psycho-pharmacological research is to create novel treatment strategies with rapid onset of the beneficial effect. The research project described in this book aimed to assess, using an animal model of depression,dynamical changes in brain neuro-chemistry and behavior following the administration of different antidepressant drugs. The main intend was to understand the neuronal mechanism of early onset of behavioral effects of antidepressant treatment. The research was focused on reward system and on serotonin-dopamine interactions in the nucleus accumbens. This book dedicated to the memory of author's devoted teacher and his primary scientific mentor, Dr. Michael E. Newman.
This chapter aims to summarize the up-today evidence-based biomedical knowledge on serotonin-2A (5-HT 2A) receptors and their role in pathophysiology and treatment of central nervous system (CNS) disorders, with a primary focus on... more
This chapter aims to summarize the up-today evidence-based biomedical knowledge on serotonin-2A (5-HT 2A) receptors and their role in pathophysiology and treatment of central nervous system (CNS) disorders, with a primary focus on depression. The first paragraph provides a brief introduction to serotonin (5-HT) system and 5-HT receptors, focusing on serotonin-2 (5-HT2) family and 5-HT2A receptor specifically. The second paragraph is focused on molecular genetics of 5-HT2A receptors, polymorphism of 5-HT2A receptor (5HT2AR) gene, 5HT2AR gene epigenetic mechanisms, such as DNA methylation, and post-translational modifications of 5-HT2AR messenger ribonucleic acid (mRNA), such as alternative splicing. The molecular and cellular pharmacology and physiology of 5-HT2A receptors in normal and pathological conditions are discussed in the third paragraph. The 5-HT2A receptors-acting ligands are addresses. The fourth paragraph describes the role of 5-HT receptors in the interaction between 5-HT and other neurotransmitter systems in health and in CNS disorders. The fifth and the final paragraph specifically deals with the role of 5-HT2A receptor in pathophysiology and treatment of depression, focusing on the 5-HT2A receptor expressed in the hippocampus.
Research Interests:
There are number of lines of evidence that the neurotransmitter norepinephrine (NE) might be very important in pathophysiology of anxiety and mood disorders. Firstly, NE projections innervate the limbic system, suggesting the involvement... more
There are number of lines of evidence that the neurotransmitter norepinephrine (NE) might be very important in pathophysiology of anxiety and mood disorders. Firstly, NE projections innervate the limbic system, suggesting the involvement of NE in the regulation of emotions and cognition. Secondly, NE interacts with serotonin (5-HT) and dopamine (DA) systems, which also play very important roles in the regulation of mood. Thirdly, it has been shown that various agents for increasing NE availability, such as NE reuptake inhibitors, are also effective antidepressant drugs. And fourthly, the depletion of NE can result in the relapse of depression after successful treatment with antidepressant drugs. All these pieces of evidence suggest that the stimulation of NE transmission can be beneficial in the treatment of affective disorders. However, different psychiatric medications have distant effects on NE transmission. The current chapter analyses the effect of psychiatric medications on NE system and proposes how the treatment outcome might be improved.
Prevalence of depression has increased progressively over the last decades. Besides the impact on human quality of life, the pharmaco-economical impact of this syndrome requires ongoing development of newer, more powerful... more
Prevalence of depression has increased progressively over the last decades. Besides the impact on human quality of life, the pharmaco-economical impact of this syndrome requires ongoing development of newer, more powerful antidepressants.  While optimizing existing therapeutic compounds, multiple approaches can be taken to generate superiority over these compounds.  The delay in onset of action of antidepressants is of relevance as the presence of side effects during the initial absence of clinical effects causes low therapy compliance.  Obviously, a decrease in onset of action would overcome this problem.  Current therapy still induces considerable side-effects depending on the class of antidepressants used.  Reducing these has multiple advantages, such as it will increase compliance but also facilitate the rapid and safe initiation of drug treatment. In line with safety requirements is the notion that new antidepressants should not be prone to hazardous effects in overdose, nor should they induce dangerous interactions by interfering with other treatment.  Finally, it is currently recognized that depression is a cluster of symptoms rather than a concise disease.  To this end, it is recognized that more tailored treatments might be required in the future. Arguably targeting sub symptoms and co-morbid features like anxiety are of high relevance.  Attempts to improve antidepressants have been made into monoamine related strategies, but also more recently in non monoamine strategies. The effectiveness of monoamine targeted selective, dual- and triple uptake inhibitors and augmented uptake inhibitors is discussed. In addition, new strategies like monoamine non-uptake inhibitor drugs or non monoamine drugs exerting effects on Glu, GABA, Substance P, and acetylcholine are discussed, as are more miscellaneous approaches.
Introduction: The current study aimed to identify and characterize the spontaneous firing activity of the neurons of rat arcuate nucleus (ARN) of the hypothalamus in vivo. The ARN contains two types of neurons: pro-opiomelanocortin (POMC)... more
Introduction: The current study aimed to identify and characterize the spontaneous firing activity of the neurons of rat arcuate nucleus (ARN) of the hypothalamus in vivo. The ARN contains two types of neurons: pro-opiomelanocortin (POMC) neurons which release β-endorphin and neuropeptide-Y (NPY) secreting neurons [1]. These neurons play an important role in pain modulation, reward, motivation, and appetite regulation. Therefore, in vivo electrophysiological characterization of the firing activity of ARN neurons might contribute to better understanding of their function under physiological and pathophysiological conditions.

Methods: Adult male and female Sprague-Dawley rats (200–250 g) were used in experiments. Rats were anesthetized with urethane (1.25 g/kg, i.p.) and mounted in stereotaxic apparatus (David Kopf, Tujunga, CA, USA). One-mm scull holes were drilled above the ARN (3.9 mm posterior and 0.6 mm lateral from the bregma). The glass electrodes filled with 2M NaCl (impedance 4–6 MΩ) were lowered throughout the lateral ARN three times in each animal (3.8, 3.9, and 4.0 mm posterior and 0.6 mm lateral from the bregma) to the depth from 9.5 to 10.0 mm ventral from the brain surface [2]. Spontaneously active neurons were recorded using HEKA-10 amplifier.

Results: Spontaneously active neurons were detected in the lateral ARN of both male and female rats. The firing pattern of the spontaneously active ARN neurons included both single action potentials and short bursts of two or three action potentials. The average duration of an action potential was ∼5 msec, with ∼1.5 msec depolarization, ∼1 msec repolarization, and ∼3 msec refractory period. The average number of spontaneously active neurons per electrode tack was slightly higher in males (2.65±0.33) than in females (1.85±0.25); however, this difference was not statistically significant. The average firing rate of spontaneous active ARN neurons was 3.14±0.50 in males (59 neurons from 7 rats) and 3.02±0.95 in females (23 neurons from 6 rats). According to preliminary results, in male rats, 75% of neurons were stimulated by intravenous administration of 1 mg/kg of leptin and 25% neurons did not respond to the leptin administration. In female rats, 50% of neurons were activated, and 25% of neurons were inhibited by leptin and 25% of neurons did not respond to the leptin administration. In summary, a significant main effect of time (Fdf17,143 = 2.45, p < 0.01) and significant interaction between gender and time (Fdf17,143 = 1.75, p < 0.05) in the response of spontaneous active ARN neurons to leptin administration was observed (two-way ANOVA for repeated measures).

Conclusion: Based on previous study [1], it can be suggested that ARN neurons which were stimulated by leptin are POMC cells. The cells which were inhibited by leptin are likely to be NPY neurons. It is possible that male Sprague-Dawley rats have higher density of spontaneously active POMC and smaller density of spontaneous active NPY neurons.

References

[1] Elias, C.F., Aschkenasi, C., Lee, C., Kelly, J., Ahima, R.S., Bjorbaek, C., Flier, J.S., Saper, C.B., Elmquist, J.K., 1999. Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron 23, 775–86.

[2] Paxinos, G., Watson, C., 2013. The Rat Brain in Stereotaxic Coordinates, Seventh Edition. Academic Press, Cambridge, UK.

Disclosure statement: This work was supported by Slovak Academy of Sciences Scholarship and by VEGA grant 2/0024/15. Leptin was provided by the National Hormone & Peptide Program (NHPP) of the University of California in Los Angeles, CA, USA.