Annamaria Lubelska
Jagiellonian University Medical College
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Annamaria Lubelska.
European Journal of Medicinal Chemistry | 2016
Katarzyna Kucwaj-Brysz; Dawid Warszycki; Sabina Podlewska; Jagna Witek; Karolina Witek; Andrea González Izquierdo; Grzegorz Satała; María Isabel Loza; Annamaria Lubelska; Gniewomir Latacz; Andrzej J. Bojarski; Marián Castro; Katarzyna Kieć-Kononowicz; Jadwiga Handzlik
A series of novel arylpiperazine 5-(4-fluorophenyl)-5-methylhydantoins with 2-hydroxypropyl linker (2-15) was synthesized and evaluated on their affinity towards serotonin 5-HT7 receptor (5-HT7R) in comparison to other closely related GPCRs: serotonin 5-HT1A, and dopamine D2 receptors. The functional activity studied through the measurement of 5-HT7R-mediated cyclic AMP production in Human Embryonic Kidney 293xa0cells (HEK293) stably expressing human 5-HT7 proved their antagonistic properties. The lead structure was also examined in the preliminary metabolic stability study using human liver microsomes (HMLs). The process of selection of candidates for synthesis was supported by a special molecular modeling workflow including combinatorial library generation, docking, and machine learning-based assessment. Additionally, in silico predictions of selectivity over 5-HT1AR and D2R, as well as functional activity were carried out. The newly synthesized compounds were proved to possess a potent affinity for 5-HT7R, similar to that of the lead structure of 5-(4-fluorophenyl)-3-(3-(4-(2-methoxyphenyl)piperazin-1-yl)-2-hydroxypropyl)-5-methylimidazolidine-2,4-dione (1). For several derivatives, significant selectivity both over 5-HT1AR and D2R was found.
Chemical Biology & Drug Design | 2017
Gniewomir Latacz; Annamaria Lubelska; Magdalena Jastrzębska-Więsek; Anna Partyka; Andrzej Sobiło; Agnieszka Olejarz; Katarzyna Kucwaj-Brysz; Grzegorz Satała; Andrzej J. Bojarski; Anna Wesołowska; Katarzyna Kieć-Kononowicz; Jadwiga Handzlik
Since the year 1993, when 5‐HT7 receptor (5‐HT7R) was discovered, there is no selective 5‐HT7R ligand introduced to the pharmaceutical market. One out of the main reasons disqualifying the 5‐HT7R ligands is weak drugability properties, including metabolic instability or low permeability. This study is focused on the search of a lead compound by “drug‐likeness” estimation of the first series of selective and potent 5‐HT7R ligands among 5‐(4‐fluorophenyl)‐3‐(2‐hydroxy‐3‐(4‐aryl‐piperazin‐1‐yl)propyl)‐5‐methylimidazolidine‐2,4‐dione derivatives (11–16). The most important drugability parameters, i.e., permeability, metabolic stability, and safety, have been evaluated. The main metabolic pathways were determined. The forced swim test (FST) in mice was performed as a primary in vivo assay for compound 13 and the reference 2. The experiments showed promising drug‐like properties for all ligands, with special attention to the benzhydryl (diphenylmethyl) derivative 13. The studies have also indicated in vivo activity of the compound 13 that was observed as a significant and specific antidepressant‐like activity in the FST. Taking into account the beneficial properties of 13, i.e., good drug‐like parameters, the significant antagonistic action, high selectivity to 5‐HT7R, and its in vivo antidepressant‐like activity, the compound should be considered as a new lead in the search for drugs acting on CNS via 5‐HT7 receptor.
Bioorganic & Medicinal Chemistry Letters | 2018
Gniewomir Latacz; Annamaria Lubelska; Magdalena Jastrzębska-Więsek; Anna Partyka; Katarzyna Kucwaj-Brysz; Anna Wesołowska; Katarzyna Kieć-Kononowicz; Jadwiga Handzlik
We report the in vitro drug-likeness studies and in vivo pharmacological evaluation for a new potent 5-HT7 receptor antagonist MF-8 (5-(4-fluorophenyl)-3-(2-hydroxy-3-(4-(2-methoxyphenyl)piperazin-1-yl)propyl)-5-methylhydantoin). The in vitro tests showed good permeability, very good metabolic stability, low risk of drug-drug interactions and satisfying safety profile. Moreover, MF-8 showed excellent antidepressant-like activity in the forced swim test in rodents and promising anxiolytic-like activity in the four-plate test in mice. Regarding the potent affinity, high selectivity and antagonistic activity of MF-8 for the 5-HT7 receptor as well as excellent drug - like properties in vitro and confirmed in vivo pharmacological activity, MF-8 should be considered as a very significant molecule in the search for a new class of anti-depressant drugs.
Journal of Medicinal Chemistry | 2018
Tommaso Felicetti; Rolando Cannalire; Donatella Pietrella; Gniewomir Latacz; Annamaria Lubelska; Giuseppe Manfroni; Maria Letizia Barreca; Serena Massari; Oriana Tabarrini; Katarzyna Kieć-Kononowicz; Bryan D. Schindler; Glenn W. Kaatz; Violetta Cecchetti; Stefano Sabatini
Antimicrobial resistance (AMR) represents a hot topic in drug discovery. Besides the identification of new antibiotics, the use of nonantibiotic molecules to block resistance mechanisms is a powerful alternative. Bacterial efflux pumps exert an early step in AMR development by allowing bacteria to grow at subinhibitorial drug concentrations. Thus, efflux pump inhibitors (EPIs) offer a great opportunity to fight AMR. Given our experience in developing Staphylococcus aureus NorA EPIs, in this work, starting from the 2-phenylquinoline hit 1, we planned the introduction of methoxy groups on the basis of their presence in known NorA EPIs. Among the 35 different synthesized derivatives, compounds 3b and 7d exhibited the best NorA inhibition activity by restoring at very low concentrations ciprofloxacin MICs against resistant S. aureus strains. Interestingly, both compounds displayed EPI activities at nontoxic concentrations for human cells as well as highlighted promising results by preliminary pharmacokinetic studies.
Pharmacology, Biochemistry and Behavior | 2018
Alaa Alachkar; Gniewomir Latacz; Agata Siwek; Annamaria Lubelska; Ewelina Honkisz; Anna Gryboś; Dorota Łażewska; Jadwiga Handzlik; Holger Stark; Katarzyna Kieć-Kononowicz; Bassem Sadek
ABSTRACT Novel non‐imidazole histamine H3 receptor (H3R) antagonists (2–8) were developed and assessed for in‐vitro antagonist binding affinities at the human histamine H1–H4R. These novel H3R antagonists (2–8) were examined in‐vivo for anticonvulsant effects in three different convulsion models in male adult rats. Compound 6 significantly and dose‐dependently exhibited decreased duration of tonic hind limb extension (THLE) in the maximal electroshock (MES)‐ and fully protected animals against pentylenetetrazole (PTZ)‐induced convulsion, following acute systemic administration (5, 10, and 20 mg/kg, i.p.). Contrary, all compounds 2–8 showed moderate protection in the strychnine (STR)‐induced convulsion model following acute pretreatment (10 mg/kg, i.p.). Moreover, the acute systemic administration of H3R antagonist 6 (10 mg/kg, i.p.) significantly prolonged latency time for MES convulsions. Furthermore, the anticonvulsant effect observed with compound 6 in MES‐model was entirely abrogated when rats were co‐injected with the brain penetrant H1R antagonist pyrilamine (PYR) but not the brain penetrant H2R antagonist zolantidine (ZOL). However, PYR and ZOL failed to abolish the full protection provided by the H3R antagonist 6 in PTZ‐ and STR‐models. No mutagenic or antiproliferative effects or potential metabolic interactions were shown for compound 6 when assessing its antiproliferative activities and metabolic profiling applying in‐vitro methods. These findings demonstrate the potential of non‐imidazole H3R antagonists as novel antiepileptic drugs (AEDs) either for single use or in addition to currently available epilepsy medications. HighlightsThe effects of novel H3R antagonists on seizure models were tested.Acute administration of H3R antagonist 6 provided promising protection in seizure models.The results shed light on the potential therapeutic importance of H3Rs in epilepsy.
Molecules | 2018
Rafał Kurczab; Wesam Ali; Dorota Łażewska; Magdalena Kotańska; Magdalena Jastrzębska-Więsek; Grzegorz Satała; Małgorzata Więcek; Annamaria Lubelska; Gniewomir Latacz; Anna Partyka; Małgorzata Starek; Monika Dąbrowska; Anna Wesołowska; Claus Jacob; Katarzyna Kieć-Kononowicz; Jadwiga Handzlik
This study focuses on the design, synthesis, biological evaluation, and computer-aided structure-activity relationship (SAR) analysis for a novel group of aromatic triazine-methylpiperazines, with an hydantoin spacer between 1,3,5-traizine and the aromatic fragment. New compounds were synthesized and their affinities for serotonin 5-HT6, 5-HT1A, 5-HT2A, 5-HT7, and dopamine D2 receptors were evaluated. The induced-fit docking (IFD) procedure was performed to explore the 5-HT6 receptor conformation space employing two lead structures. It resulted in a consistent binding mode with the activity data. For the most active compounds found in each modification line, anti-obesity and anti-depressive-like activity in vivo, as well as “druglikeness” in vitro, were examined. Two 2-naphthyl compounds (18 and 26) were identified as the most active 5-HT6R agents within each lead modification line, respectively. The 5-(2-naphthyl)hydantoin derivative 26, the most active one in the series (5-HT6R: Ki = 87 nM), displayed also significant selectivity towards competitive G-protein coupled receptors (6–197-fold). Docking studies indicated that the hydantoin ring is stabilized by hydrogen bonding, but due to its different orientation, the hydrogen bonds form with S5.44 and N6.55 or Q6.58 for 18 and 26, respectively. Compound 26 exerted anxiolytic-like and antidepressant-like activities. Importantly, it demonstrated anti-obesity properties in animals fed palatable feed, and did not show toxic effects in vitro.
European Journal of Medicinal Chemistry | 2018
Katarzyna Szczepańska; Tadeusz Karcz; Szczepan Mogilski; Agata Siwek; Kamil Kuder; Gniewomir Latacz; Monika Kubacka; Stefanie Hagenow; Annamaria Lubelska; Agnieszka Olejarz; Magdalena Kotańska; Bassem Sadek; Holger Stark; Katarzyna Kieć-Kononowicz
As a continuation of our search for novel histamine H3 receptor ligands, a series of twenty four new tert-butyl and tert-pentyl phenoxyalkylamine derivatives (2-25) was synthesized. Compounds with three to four carbon atoms alkyl chain spacer were evaluated for their binding properties at human histamine H3 receptor (hH3R). The highest affinities were observed for 4-pyridyl derivatives 4, 10, 16 and 22 (Kiu202f=u202f16.0-120u202fnM). As it has been shown in docking studies, those specific heteroaromatic 4-N piperazine substituents might interact with one of the key receptor interacting amino acids. Moreover, the most promising compounds exhibited anticonvulsant activity in the maximal electroshock-induced seizure (MES) model in mice. Furthermore, the blood-brain barrier penetration, the functional H3R antagonist potency as well as the pro-cognitive properties in the passive avoidance test were demonstrated for compound 10. In order to estimate drug-likeness of compound 10,in silico and experimental evaluation of metabolic stability in human liver microsomes was performed. In addition, paying attention to the results obtained within this study, the 4-pyridyl-piperazino moiety has been established as a new bioisosteric piperidine replacement in H3R ligands.
Bioorganic & Medicinal Chemistry | 2018
Dorota Łażewska; Maria Kaleta; Stefanie Hagenow; Szczepan Mogilski; Gniewomir Latacz; Tadeusz Karcz; Annamaria Lubelska; Ewelina Honkisz; Jadwiga Handzlik; David Reiner; Grzegorz Satała; Barbara Filipek; Holger Stark; Katarzyna Kieć-Kononowicz
A series of 1- and 2-naphthyloxy derivatives were synthesized and evaluated for histamine H3 receptor affinity. Most compounds showed high affinities with Ki values below 100u202fnM. The most potent ligand, 1-(5-(naphthalen-1-yloxy)pentyl)azepane (11) displayed high affinity for the histamine H3 receptor with a Ki value of 21.9u202fnM. The antagonist behaviour of 11 was confirmed both in vitro in the cAMP assay (IC50u202f=u202f312u202fnM) and in vivo in the rat dipsogenia model (ED50u202f=u202f3.68u202fnM). Moreover, compound 11 showed positive effects on scopolamine induced-memory deficits in mice (at doses of 10 and 15u202fmg/kg) and an analgesic effect in the formalin test in mice with ED50u202f=u202f30.6u202fmg/kg (early phase) and ED50u202f=u202f20.8u202fmg/kg (late phase). Another interesting compound, 1-(5-(Naphthalen-1-yloxy)pentyl)piperidine (13; H3R Kiu202f=u202f53.9u202fnM), was accepted for Anticonvulsant Screening Program at the National Institute of Neurological Disorders and Stroke/National Institute of Health (Rockville, USA). The screening was performed in the maximal electroshock seizure (MES), the subcutaneous pentylenetetrazole (scPTZ) and the 6-Hz psychomotor animal models of epilepsy. Neurologic deficit was evaluated by the rotarod test. Compound 13 inhibited convulsions induced by the MES with ED50 of 19.2u202fmg/kg (mice, i.p.), 17.8 (rats, i.p.), and 78.1 (rats, p.o.). Moreover, 13 displayed protection against the 6-Hz psychomotor seizures (32u202fmA) in mice (i.p.) with ED50 of 33.1u202fmg/kg and (44u202fmA) ED50 of 57.2u202fmg/kg. Furthermore, compounds 11 and 13 showed in vitro weak influence on viability of tested cell lines (normal HEK293, neuroblastoma IMR-32, hepatoma HEPG2), weak inhibition of CYP3A4 activity, and no mutagenicity. Thus, these compounds may be used as leads in a further search for histamine H3 receptor ligands with promising in vitro and in vivo activity.
ACS Chemical Neuroscience | 2018
Katarzyna Socała; Szczepan Mogilski; Mateusz Pieróg; Dorota Nieoczym; Michał Abram; Bartłomiej Szulczyk; Annamaria Lubelska; Gniewomir Latacz; Urszula Doboszewska; Piotr Wlaź; Krzysztof Kamiński
Recently, compound KA-11 was identified as a promising candidate for a new broad-spectrum anticonvulsant. This compound revealed wide protective activity across the most important animal models of seizures such as the maximal electroshock test (MES), the subcutaneous pentylenetetrazole test ( scPTZ), and the six-hertz test (6 Hz, 32 mA). Importantly, KA-11 was devoid of acute neurological activity, which was assessed by applying the chimney test (TD50 value higher than 1500 mg/kg). The preliminary in vivo results confirmed favorable anticonvulsant and safety properties of KA-11. With the aim of further biological characterization of KA-11, in the current studies we evaluated its antiepileptogenic activity in the kindling model of epilepsy induced by repeated injection of PTZ in mice. Furthermore, we assessed the antinociceptive activity of KA-11 in several animal pain models. As a result, KA-11 (at all doses applied: 25, 50, and 100 mg/kg) significantly delayed the progression of kindling induced by repeated injection of PTZ in mice. Additionally, KA-11 revealed potent antinociceptive activity in the formalin-induced tonic pain and, importantly, in the oxaliplatin-induced neuropathic pain model in mice. Moreover, KA-11 did not induce motor deficits in the rotarod test. Patch-clamp experiments revealed that one of the mechanisms of action of KA-11 is inhibition of voltage-gated sodium currents. Compound KA-11 appeared to be safe in relation to hepatotoxic properties as no phospholipidosis induction was determined in HepG2 cells at 50 μM, and a small, statistically significant decrease of cell viability was observed only at the highest used dose of 100 μM. Moreover, KA-11 did not affect the function of CYP2D6. The aforementioned hybrid substance proved to penetrate the biological membranes in the in vitro permeability assays.
Bioorganic & Medicinal Chemistry | 2017
Dorota Łażewska; Maria Kaleta; J. Stephan Schwed; Tadeusz Karcz; Szczepan Mogilski; Gniewomir Latacz; Agnieszka Olejarz; Agata Siwek; Monika Kubacka; Annamaria Lubelska; Ewelina Honkisz; Jadwiga Handzlik; Barbara Filipek; Holger Stark; Katarzyna Kieć-Kononowicz
Novel biphenyloxy-alkyl derivatives of piperidine and azepane were synthesized and evaluated for their binding properties at the human histamine H3 receptor. Two series of compounds were obtained with a meta- and a para-biphenyl moiety. The alkyl chain spacer contained five and six carbon atoms. The highest affinity among all compounds was shown by 1-(6-(3-phenylphenoxy)hexyl)azepane (13) with a Ki value of 18nM. Two para-biphenyl derivatives, 1-(5-(4-phenylphenoxy)pentyl)piperidine (14; Ki=25nM) and 1-(5-(4-phenylphenoxy)pentyl)azepane (16; Ki=34nM), classified as antagonists in a cAMP accumulation assay (IC50=4 and 9nM, respectively), were studied in detail. Compounds 14 and 16 blocked RAMH-induced dipsogenia in rats (ED50 of 2.72mg/kg and 1.75mg/kg respectively), and showed high selectivity (hH4R vs hH3R>600-fold) and low toxicity (hERG inhibition: IC50>1.70µM; hepatotoxicity IC50>12.5µM; non-mutagenic up to 10µM). Furthermore, the metabolic stability was evaluated in vitro on human liver microsomes (HLMs) and/or rat liver microsomes (RLMs). Metabolites produced were analyzed and tentatively identified by UPLC-MS techniques. The results demonstrated easy hydroxylation of the biphenyl ring.