1
|
Witkowski G, Szulczyk B, Nurowska E, Jurek M, Pasierski M, Lipiec A, Charzewska A, Dawidziuk M, Milewski M, Owsiak S, Rola R, Sienkiewicz Jarosz H, Hoffman-Zacharska D. Functional Characteristics of the Nav1.1 p.Arg1596Cys Mutation Associated with Varying Severity of Epilepsy Phenotypes. Int J Mol Sci 2024; 25:1745. [PMID: 38339022 PMCID: PMC10855957 DOI: 10.3390/ijms25031745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Mutations of the SCN1A gene, which encodes the voltage-dependent Na+ channel's α subunit, are associated with diverse epileptic syndromes ranging in severity, even intra-family, from febrile seizures to epileptic encephalopathy. The underlying cause of this variability is unknown, suggesting the involvement of additional factors. The aim of our study was to describe the properties of mutated channels and investigate genetic causes for clinical syndromes' variability in the family of five SCN1A gene p.Arg1596Cys mutation carriers. The analysis of additional genetic factors influencing SCN1A-associated phenotypes was conducted through exome sequencing (WES). To assess the impact of mutations, we used patch clamp analysis of mutated channels expressed in HEK cells and in vivo neural excitability studies (NESs). In cells expressing the mutant channel, sodium currents were reduced. NESs indicated increased excitability of peripheral motor neurons in mutation carriers. WES showed the absence of non-SCA1 pathogenic variants that could be causative of disease in the family. Variants of uncertain significance in three genes, as potential modifiers of the most severe phenotype, were identified. The p.Arg1596Cys substitution inhibits channel function, affecting steady-state inactivation kinetics. Its clinical manifestations involve not only epileptic symptoms but also increased excitability of peripheral motor fibers. The role of Nav1.1 in excitatory neurons cannot be ruled out as a significant factor of the clinical phenotype.
Collapse
Affiliation(s)
- Grzegorz Witkowski
- First Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland; (S.O.); (H.S.J.)
- Military Institute of Aviation Medicine, Krasinskiego 54/56, 01-755 Warsaw, Poland;
| | - Bartlomiej Szulczyk
- Chair and Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, The Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.S.); (E.N.); (M.P.)
| | - Ewa Nurowska
- Chair and Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, The Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.S.); (E.N.); (M.P.)
| | - Marta Jurek
- Department of Medical Genetics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (M.J.); (A.C.); (M.M.); (D.H.-Z.)
| | - Michal Pasierski
- Chair and Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, The Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.S.); (E.N.); (M.P.)
| | - Agata Lipiec
- Clinic of Pediatric Neurology, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland;
| | - Agnieszka Charzewska
- Department of Medical Genetics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (M.J.); (A.C.); (M.M.); (D.H.-Z.)
| | - Mateusz Dawidziuk
- Department of Medical Genetics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (M.J.); (A.C.); (M.M.); (D.H.-Z.)
| | - Michal Milewski
- Department of Medical Genetics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (M.J.); (A.C.); (M.M.); (D.H.-Z.)
| | - Szymon Owsiak
- First Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland; (S.O.); (H.S.J.)
| | - Rafal Rola
- Military Institute of Aviation Medicine, Krasinskiego 54/56, 01-755 Warsaw, Poland;
| | - Halina Sienkiewicz Jarosz
- First Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland; (S.O.); (H.S.J.)
| | - Dorota Hoffman-Zacharska
- Department of Medical Genetics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (M.J.); (A.C.); (M.M.); (D.H.-Z.)
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| |
Collapse
|
2
|
Stöber TM, Batulin D, Triesch J, Narayanan R, Jedlicka P. Degeneracy in epilepsy: multiple routes to hyperexcitable brain circuits and their repair. Commun Biol 2023; 6:479. [PMID: 37137938 PMCID: PMC10156698 DOI: 10.1038/s42003-023-04823-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 04/06/2023] [Indexed: 05/05/2023] Open
Abstract
Due to its complex and multifaceted nature, developing effective treatments for epilepsy is still a major challenge. To deal with this complexity we introduce the concept of degeneracy to the field of epilepsy research: the ability of disparate elements to cause an analogous function or malfunction. Here, we review examples of epilepsy-related degeneracy at multiple levels of brain organisation, ranging from the cellular to the network and systems level. Based on these insights, we outline new multiscale and population modelling approaches to disentangle the complex web of interactions underlying epilepsy and to design personalised multitarget therapies.
Collapse
Affiliation(s)
- Tristan Manfred Stöber
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, 44801, Bochum, Germany
- Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Goethe University, 60590, Frankfurt, Germany
| | - Danylo Batulin
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
- CePTER - Center for Personalized Translational Epilepsy Research, Goethe University, 60590, Frankfurt, Germany
- Faculty of Computer Science and Mathematics, Goethe University, 60486, Frankfurt, Germany
| | - Jochen Triesch
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Peter Jedlicka
- ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus Liebig University Giessen, 35390, Giessen, Germany.
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
3
|
Kuanyshbek A, Wang M, Andersson Å, Tuifua M, Palmer EE, Sachdev RK, Mu TW, Vetter I, Keramidas A. Anti-seizure mechanisms of midazolam and valproate at the β2(L51M) variant of the GABA A receptor. Neuropharmacology 2022; 221:109295. [PMID: 36257447 PMCID: PMC9981329 DOI: 10.1016/j.neuropharm.2022.109295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
Abstract
Genetic sequencing is identifying an expanding number of variants of GABAA receptors associated with human epilepsies. We identified a new de novo variant of the β2 subunit (β2L51M) of the inhibitory GABAA receptor associated with seizures. Our analysis determined the pathogenicity of the variant and the effects of anti-seizure medications. Our data demonstrates that the variant reduced cell surface trafficking and peak GABA-gated currents. Synaptic currents mediated by variant-containing receptors decayed faster than wild-type and single receptor currents showed that the variant shortened the duration of receptor activity by decreasing receptor open times. We tested the effects of the anti-seizure medications, midazolam, carbamazepine and valproate and found that all three enhance variant receptor surface expression. Additionally, midazolam restored receptor function by increasing single receptor active periods and synaptic current decay times towards wild-type levels. By contrast, valproate increased synaptic peak currents, event frequency and promoted synaptic bursting. Our study identifies a new disease-causing variant to the GABAA receptor, profiles its pathogenic effects and demonstrates how anti-seizure drugs correct its functional deficits.
Collapse
Affiliation(s)
- Alibek Kuanyshbek
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Meng Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Åsa Andersson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Marie Tuifua
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Elizabeth E Palmer
- Sydney Children's Hospital Network, Randwick Sydney Australia and School of Women's and Children's Health, UNSW Medicine, The University of New South Wales, Sydney, NSW, Australia
| | - Rani K Sachdev
- Sydney Children's Hospital Network, Randwick Sydney Australia and School of Women's and Children's Health, UNSW Medicine, The University of New South Wales, Sydney, NSW, Australia
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, 4102, Australia
| | - Angelo Keramidas
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
4
|
Abram M, Jakubiec M, Rapacz A, Mogilski S, Latacz G, Szulczyk B, Szafarz M, Socała K, Nieoczym D, Wyska E, Wlaź P, Kamiński RM, Kamiński K. Identification of New Compounds with Anticonvulsant and Antinociceptive Properties in a Group of 3-substituted (2,5-dioxo-pyrrolidin-1-yl)(phenyl)-Acetamides. Int J Mol Sci 2021; 22:ijms222313092. [PMID: 34884898 PMCID: PMC8658016 DOI: 10.3390/ijms222313092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 12/30/2022] Open
Abstract
We report herein a series of water-soluble analogues of previously described anticonvulsants and their detailed in vivo and in vitro characterization. The majority of these compounds demonstrated broad-spectrum anticonvulsant properties in animal seizure models, including the maximal electroshock (MES) test, the pentylenetetrazole-induced seizure model (scPTZ), and the psychomotor 6 Hz (32 mA) seizure model in mice. Compound 14 showed the most robust anticonvulsant activity (ED50 MES = 49.6 mg/kg, ED50 6 Hz (32 mA) = 31.3 mg/kg, ED50scPTZ = 67.4 mg/kg). Notably, it was also effective in the 6 Hz (44 mA) model of drug-resistant epilepsy (ED50 = 63.2 mg/kg). Apart from favorable anticonvulsant properties, compound 14 revealed a high efficacy against pain responses in the formalin-induced tonic pain, the capsaicin-induced neurogenic pain, as well as in the oxaliplatin-induced neuropathic pain in mice. Moreover, compound 14 showed distinct anti-inflammatory activity in the model of carrageenan-induced aseptic inflammation. The mechanism of action of compound 14 is likely complex and may result from the inhibition of peripheral and central sodium and calcium currents, as well as the TRPV1 receptor antagonism as observed in the in vitro studies. This lead compound also revealed beneficial in vitro ADME-Tox properties and an in vivo pharmacokinetic profile, making it a potential candidate for future preclinical development. Interestingly, the in vitro studies also showed a favorable induction effect of compound 14 on the viability of neuroblastoma SH-SY5Y cells.
Collapse
Affiliation(s)
- Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (M.A.); (M.J.); (R.M.K.)
| | - Marcin Jakubiec
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (M.A.); (M.J.); (R.M.K.)
| | - Anna Rapacz
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (A.R.); (S.M.)
| | - Szczepan Mogilski
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (A.R.); (S.M.)
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland;
| | - Bartłomiej Szulczyk
- Department of Pharmacodynamics, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
| | - Małgorzata Szafarz
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (M.S.); (E.W.)
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (K.S.); (D.N.); (P.W.)
| | - Dorota Nieoczym
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (K.S.); (D.N.); (P.W.)
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (M.S.); (E.W.)
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (K.S.); (D.N.); (P.W.)
| | - Rafał M. Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (M.A.); (M.J.); (R.M.K.)
| | - Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (M.A.); (M.J.); (R.M.K.)
- Correspondence: ; Tel.: +48-12-620-54-59
| |
Collapse
|
5
|
Szulczyk B, Pasierski M, Gawlak M. Prefrontal cortex pyramidal neurons express functional Nav1.8 tetrodotoxin-resistant sodium currents. Clin Exp Pharmacol Physiol 2021; 49:350-359. [PMID: 34750860 DOI: 10.1111/1440-1681.13610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 11/27/2022]
Abstract
It has been repeatedly proved that Nav1.8 tetrodotoxin (TTX)-resistant sodium currents are expressed in peripheral sensory neurons where they play important role in nociception. There are very few publications that show the presence of TTX-resistant sodium currents in central neurons. The aim of this study was to assess if functional Nav1.8 TTX-resistant sodium currents are expressed in prefrontal cortex pyramidal neurons. All recordings were performed in the presence of TTX in the extracellular solution to block TTX-sensitive sodium currents. The TTX-resistant sodium current recorded in this study was mainly carried by the Nav1.8 sodium channel isoform because the Nav1.9 current was inhibited by the -65 mV holding potential that we used throughout the study. Moreover, the sodium current that we recorded was inhibited by treatment with the selective Nav1.8 inhibitor A-803467. Confocal microscopy experiments confirmed the presence of the Nav1.8 α subunit in prefrontal cortex pyramidal neurons. Activation and steady state inactivation properties of TTX-resistant sodium currents were also assessed in this study and they were similar to activation and inactivation properties of TTX-resistant sodium currents expressed in dorsal root ganglia (DRG) neurons. Moreover, this study showed that carbamazepine (60 µM) inhibited the maximal amplitude of the TTX-resistant sodium current. Furthermore, we found that carbamazepine shifts steady state inactivation curve of TTX-resistant sodium currents toward hyperpolarization. This study suggests that the Nav1.8 TTX-resistant sodium channel is expressed not only in DRG neurons, but also in cortical neurons and may be molecular target for antiepileptic drugs such as carbamazepine.
Collapse
Affiliation(s)
- Bartłomiej Szulczyk
- Department of Pharmacodynamics, The Medical University of Warsaw, Warsaw, Poland
| | - Michał Pasierski
- Department of Pharmacodynamics, The Medical University of Warsaw, Warsaw, Poland
| | - Maciej Gawlak
- Department of Pharmacodynamics, The Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
6
|
Asymmetric synthesis and in vivo/in vitro characterization of new hybrid anticonvulsants derived from (2,5-dioxopyrrolidin-1-yl)phenylacetamides. Bioorg Chem 2021; 109:104751. [PMID: 33647745 DOI: 10.1016/j.bioorg.2021.104751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 11/21/2022]
Abstract
In the current studies we carried out an optimized multistep asymmetric synthesis of R-enantiomers (eutomers) for a previously identified series of racemic hybrid anticonvulsants. The spatial structure of selected enantiomers was solved by the use of crystallographic methods. The compound (R)-16 was identified as a lead, which revealed broad-spectrum protective activity in a range of epilepsy models with the following ED50 values: the maximal electroshock (MES) test (36.0 mg/kg), the 6 Hz (32 mA) seizure model (39.2 mg/kg), and the pentylenetetrazole-induced seizure model (scPTZ) (54.8 mg/kg). Furthermore, (R)-16 displayed a low potency for the induction of motor impairment in the rotarod test (TD50 = 468.5 mg/kg), resulting in potentially very beneficial therapeutic window. Finally, (R)-16 showed satisfying ADME-Tox properties in the in vitro assays. Therefore, the data obtained in the current studies justify the further preclinical development of (R)-16 as candidate for potentially broad-spectrum and safe anticonvulsant.
Collapse
|
7
|
Kamiński K, Mogilski S, Abram M, Rapacz A, Latacz G, Szulczyk B, Walczak M, Kuś K, Matyjaszczyk K, Kamiński RM. KA-104, a new multitargeted anticonvulsant with potent antinociceptive activity in preclinical models. Epilepsia 2020; 61:2119-2128. [PMID: 32929733 DOI: 10.1111/epi.16669] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 01/02/2023]
Abstract
OBJECTIVE The main objective of the present work was to assess the utility of KA-104 as potential therapy for drug-resistant seizures and neuropathic pain, and to characterize its druglike properties in a series of absorption, distribution, metabolism, excretion and toxicity (ADME-Tox) studies. We also aimed to establish its mechanism of action in electrophysiological studies. METHODS The activity of KA-104 against drug-resistant seizures was tested in the mouse 6-Hz (44-mA) model, whereas the antinociceptive activity was assessed with the capsaicin- and oxaliplatin-induced pain models in mice. The patch-clamp technique was used to study the influence of KA-104 on fast voltage-gated sodium currents in rat prefrontal cortex pyramidal neurons. The pharmacokinetic profile was determined after intraperitoneal (ip) injection in mice. The in vitro ADME-Tox properties were studied by applying routine testing procedures. RESULTS KA-104 was effective in the 6-Hz (44-mA) model (median effective dose [ED50 ] = 73.2 mg/kg) and revealed high efficacy in capsaicin-induced neurogenic pain as well as in oxaliplatin-induced neuropathic pain in mice. Patch-clamp technique showed that KA-104 reversibly inhibits voltage-gated sodium currents. KA-104 was rapidly absorbed after the ip injection and showed relatively good penetration through the blood-brain barrier. This molecule was also characterized by high passive permeability, moderate influence on CYP2C9, and negligible hepatotoxicity on HepG2 cells. SIGNIFICANCE The results reported herein indicate that KA-104 is a new wide-spectrum multitargeted anticonvulsant with favorable in vitro ADME-Tox properties. Importantly, this compound may also prove to become an interesting and hopefully more effective therapeutic option for treatment of neuropathic pain.
Collapse
Affiliation(s)
- Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland
| | - Szczepan Mogilski
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland
| | - Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland
| | - Anna Rapacz
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland
| | - Bartłomiej Szulczyk
- Department of Pharmacodynamics, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Maria Walczak
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland.,Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Cracow, Poland
| | - Kamil Kuś
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland.,Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Cracow, Poland
| | - Karolina Matyjaszczyk
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland.,Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Cracow, Poland
| | - Rafał M Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
8
|
Kamiński K, Socała K, Zagaja M, Andres-Mach M, Abram M, Jakubiec M, Pieróg M, Nieoczym D, Rapacz A, Gawel K, Esguerra CV, Latacz G, Lubelska A, Szulczyk B, Szewczyk A, Łuszczki JJ, Wlaź P. N-Benzyl-(2,5-dioxopyrrolidin-1-yl)propanamide (AS-1) with Hybrid Structure as a Candidate for a Broad-Spectrum Antiepileptic Drug. Neurotherapeutics 2020; 17:309-328. [PMID: 31486023 PMCID: PMC7007424 DOI: 10.1007/s13311-019-00773-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In our recent studies, we identified compound N-benzyl-2-(2,5-dioxopyrrolidin-1-yl)propanamide (AS-1) as a broad-spectrum hybrid anticonvulsant which showed potent protection across the most important animal acute seizure models such as the maximal electroshock (MES) test, the subcutaneous pentylenetetrazole (s.c. PTZ) test, and the 6-Hz (32 mA) test in mice. Therefore, AS-1 may be recognized as a candidate for new anticonvulsant effective in different types of human epilepsy with a favorable safety margin profile determined in the rotarod test in mice. In the aim of further pharmacological evaluation of AS-1, in the current study, we examined its activity in the 6-Hz (44 mA) test, which is known as the model of drug-resistant epilepsy. Furthermore, we determined also the antiseizure activity in the kindling model of epilepsy induced by repeated injection of pentylenetetrazole (PTZ) in mice. As a result, AS-1 revealed relatively potent protection in the 6-Hz (44 mA) test, as well as delayed the progression of kindling induced by repeated injection of PTZ in mice at doses of 15 mg/kg, 30 mg/kg, and 60 mg/kg. Importantly, the isobolographic analysis showed that a combination of AS-1 and valproic acid (VPA) at the fixed ratio of 1:1 displayed a supra-additive (synergistic) interaction against PTZ-induced seizures in mice. Thus, AS-1 may be potentially used in an add-on therapy with VPA. Moreover, incubation of zebrafish larvae with AS-1 substantially decreased the number, cumulative but not the mean duration of epileptiform-like events in electroencephalographic assay. Finally, the in vitro ADME-Tox studies revealed that AS-1 is characterized by a very good permeability in the parallel artificial membrane permeability assay test, excellent metabolic stability on human liver microsomes (HLMs), no significant influence on CYP3A4/CYP2D6 activity, and moderate inhibition of CYP2C9 in a concentration of 10 μM, as well as no hepatotoxic properties in HepG2 cells (concentration of 10 μM).
Collapse
Affiliation(s)
- Krzysztof Kamiński
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, Medyczna 9, 30-688, Cracow, Poland
| | - Katarzyna Socała
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Mirosław Zagaja
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090, Lublin, Poland
| | - Marta Andres-Mach
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090, Lublin, Poland
| | - Michał Abram
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, Medyczna 9, 30-688, Cracow, Poland
| | - Marcin Jakubiec
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, Medyczna 9, 30-688, Cracow, Poland
| | - Mateusz Pieróg
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Dorota Nieoczym
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Anna Rapacz
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacodynamics, Medyczna 9, 30-688, Cracow, Poland
| | - Kinga Gawel
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Camila V Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
| | - Gniewomir Latacz
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Medyczna 9, 30-688, Cracow, Poland
| | - Annamaria Lubelska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Medyczna 9, 30-688, Cracow, Poland
| | - Bartłomiej Szulczyk
- Department of Drug Technology and Pharmaceutical Biotechnology, Medical University of Warsaw, Banacha 1, 02-097, Warsaw, Poland
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Aleksandra Szewczyk
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090, Lublin, Poland
| | - Jarogniew Jacek Łuszczki
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090, Lublin, Poland
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Piotr Wlaź
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| |
Collapse
|
9
|
Valproic acid interactions with the NavMs voltage-gated sodium channel. Proc Natl Acad Sci U S A 2019; 116:26549-26554. [PMID: 31822620 DOI: 10.1073/pnas.1909696116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Valproic acid (VPA) is an anticonvulsant drug that is also used to treat migraines and bipolar disorder. Its proposed biological targets include human voltage-gated sodium channels, among other membrane proteins. We used the prokaryotic NavMs sodium channel, which has been shown to be a good exemplar for drug binding to human sodium channels, to examine the structural and functional interactions of VPA. Thermal melt synchrotron radiation circular dichroism spectroscopic binding studies of the full-length NavMs channel (which includes both pore and voltage sensor domains), and a pore-only construct, undertaken in the presence and absence of VPA, indicated that the drug binds to and destabilizes the channel, but not the pore-only construct. This is in contrast to other antiepileptic compounds that have previously been shown to bind in the central hydrophobic core of the pore region of the channel, and that tend to increase the thermal stability of both pore-only constructs and full-length channels. Molecular docking studies also indicated that the VPA binding site is associated with the voltage sensor, rather than the hydrophobic cavity of the pore domain. Electrophysiological studies show that VPA influences the block and inactivation rates of the NavMs channel, although with lower efficacy than classical channel-blocking compounds. It thus appears that, while VPA is capable of binding to these voltage-gated sodium channels, it has a very different mode and site of action than other anticonvulsant compounds.
Collapse
|
10
|
Valproic acid potently inhibits interictal-like epileptiform activity in prefrontal cortex pyramidal neurons. Neurosci Lett 2019; 708:134350. [PMID: 31247226 DOI: 10.1016/j.neulet.2019.134350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/08/2019] [Accepted: 06/22/2019] [Indexed: 01/20/2023]
Abstract
Valproic acid has a long-standing reputation of effectively treating the symptoms of not only epilepsy but also psychiatric conditions. In the latter, the exact mechanism by which valproate exerts its effect remains unclear. In this study, epileptiform bursts were recorded from pyramidal neurons in the prefrontal cortex (the brain region thought to be involved in psychiatric disorders) using the patch-clamp technique. An extracellular solution with no magnesium ions and elevated potassium levels that is known to induce epileptiform activity in vitro was used. Because of their short durations, the epileptiform bursts were regarded as interictal-like epileptiform activity, which is believed to be involved in cognitive impairment. Interictal discharges occur in many neuropsychiatric disorders as well as in healthy population. Epileptic activity in prefrontal cortex pyramidal neurons was potently inhibited by two therapeutic concentrations of valproic acid (20 μM and 200 μM). Moreover, valproate suppressed spontaneous excitatory postsynaptic potentials. Epileptiform bursts were fully inhibited by NMDA receptor antagonist, which suggests that epileptiform activity is driven by NMDA receptors. The inhibition of excitability in prefrontal cortex pyramidal neurons by valproate was also shown. This study shows that it is possible to evoke NMDA-dependent epileptiform activity in prefrontal cortex pyramidal neurons in vitro. We suggest that the prefrontal cortex is a good region for studying the influence of drugs on interictal epileptiform activity.
Collapse
|
11
|
KCNQ2 related early-onset epileptic encephalopathies in Chinese children. J Neurol 2019; 266:2224-2232. [DOI: 10.1007/s00415-019-09404-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/23/2019] [Accepted: 05/25/2019] [Indexed: 12/20/2022]
|
12
|
Shi L, Zhu M, Li H, Wen Z, Chen X, Luo J, Lin C, Zhang Z. SCN1A and SCN2A polymorphisms are associated with response to valproic acid in Chinese epilepsy patients. Eur J Clin Pharmacol 2019; 75:655-663. [PMID: 30693367 DOI: 10.1007/s00228-019-02633-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/16/2019] [Indexed: 01/03/2023]
Abstract
PURPOSE There is a large inter-individual variation in the efficacy of valproic acid (VPA) against epilepsy. The genetic polymorphism influence of sodium channels on VPA response remains a matter of debate. The aim of the study was to explore the effect of SCN1A and SCN2A gene polymorphisms on VPA response in the treatment of epilepsy among Chinese patients. METHODS A total of 354 epileptic patients with VPA treatment were genotyped for five single nucleotide polymorphisms (SNP), including SCN1A rs10188577 T>C, rs2298771 T>C, rs3812718 G>A, and SCN2A rs2304016 A>G, rs17183814 G>A. A binary logistic regression analysis was performed to evaluate the association of genotype with VPA antiepileptic effects, adjusting the influence of confounding factors. RESULTS Genotype distributions of all selected SNPs were consistent with the Hardy-Weinberg equilibrium in epilepsy patients. SCN1A rs3812718 and SCN2A rs2304016 were found to be significantly associated with VPA response, both in monotherapy and in VPA-based polytherapy. Patients with the rs3812718 A allele were more frequently seen in the VPA-responsive group (P < 0.05), and the rs2304016 G allele was related to an increased risk of resistance to VPA therapy (P < 0.05). CONCLUSIONS Our study revealed that SCN1A rs3812718 and SCN2A rs2304016 polymorphisms might be markers of VPA response in Chinese epilepsy patients. TRIAL REGISTRATION ChiCTR-1800016477.
Collapse
Affiliation(s)
- Lihong Shi
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.,Institute of Hospital Pharmacy, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Miaomiao Zhu
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.,Institute of Hospital Pharmacy, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Huilan Li
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.,Institute of Hospital Pharmacy, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Zhipeng Wen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, No. 110 Xiangya Road, Changsha, 410008, China.,Institute of Clinical Pharmacology, Central South University, No. 110 Xiangya Road, Changsha, 410008, China
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, No. 110 Xiangya Road, Changsha, 410008, China.,Institute of Clinical Pharmacology, Central South University, No. 110 Xiangya Road, Changsha, 410008, China
| | - Jia Luo
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.,Institute of Hospital Pharmacy, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Cong Lin
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.,Institute of Hospital Pharmacy, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Zanling Zhang
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China. .,Institute of Hospital Pharmacy, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
13
|
Socała K, Mogilski S, Pieróg M, Nieoczym D, Abram M, Szulczyk B, Lubelska A, Latacz G, Doboszewska U, Wlaź P, Kamiński K. KA-11, a Novel Pyrrolidine-2,5-dione Derived Broad-Spectrum Anticonvulsant: Its Antiepileptogenic, Antinociceptive Properties and in Vitro Characterization. ACS Chem Neurosci 2019; 10:636-648. [PMID: 30247871 DOI: 10.1021/acschemneuro.8b00476] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Katarzyna Socała
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Szczepan Mogilski
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Mateusz Pieróg
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Dorota Nieoczym
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Bartłomiej Szulczyk
- Department of Drug Technology and Pharmaceutical Biotechnology, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
- Department of Physiology and Pathophysiology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Annamaria Lubelska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Urszula Doboszewska
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Piotr Wlaź
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
14
|
Yoshizawa K, Arai N, Suzuki Y, Nakamura T, Takeuchi K, Sakamoto R, Masuda R. Evaluation of the antinociceptive activities of several sodium channel blockers using veratrine test in mice. Synapse 2018; 72. [PMID: 29993143 DOI: 10.1002/syn.22056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/11/2018] [Accepted: 06/21/2018] [Indexed: 12/19/2022]
Abstract
An important role of voltage-gated sodium channels (VGSCs) in many different pain states has been established in animal models and humans wherein sodium channel blockers partially ameliorate pain. However, behavioral tests for screening analgesics that exhibit pharmacologic action by acting on VGSCs are rarely reported, and there are no studies on antinociception using veratrine as a nociceptive agent. The aim of the present study was to examine the amount of nociceptive behavior evoked by subcutaneous administration of veratrine into the hind paw and investigate whether veratrine can be used as a VGSC agonist to test the pharmacological properties of candidate analgesics via sodium channel blockade. We report for the first time that intraplantar injection of veratrine produced a reproducible nociceptive response in mice. Furthermore, several sodium channel blockers, namely carbamazepine, valproate, mexiletine, and the selective Nav1.7 inhibitor PF-04856264, but not flecainide or pilsicainide, reduced veratrine-induced nociception. In contrast, calcium channel blockers gabapentin and ethosuximide did not change veratrine-induced nociception. The veratrine test in mice might be a useful tool, at least in part, to evaluate the potential analgesic effect of sodium channel blockers.
Collapse
Affiliation(s)
- Kazumi Yoshizawa
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Narumaki Arai
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Yukina Suzuki
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Toka Nakamura
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Kota Takeuchi
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Reinii Sakamoto
- Department of Anesthesiology, Tokai University Hachioji Hospital, Tokyo, Japan
| | - Ritsuko Masuda
- Department of Anesthesiology, Tokai University Hachioji Hospital, Tokyo, Japan
| |
Collapse
|
15
|
Gawlak M, Szulczyk B, Berłowski A, Grzelka K, Stachurska A, Pełka J, Czarzasta K, Małecki M, Kurowski P, Nurowska E, Szulczyk P. Age-dependent expression of Nav1.9 channels in medial prefrontal cortex pyramidal neurons in rats. Dev Neurobiol 2017; 77:1371-1384. [PMID: 28913981 DOI: 10.1002/dneu.22537] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 12/19/2022]
Abstract
Developmental changes that occur in the prefrontal cortex during adolescence alter behavior. These behavioral alterations likely stem from changes in prefrontal cortex neuronal activity, which may depend on the properties and expression of ion channels. Nav1.9 sodium channels conduct a Na+ current that is TTX resistant with a low threshold and noninactivating over time. The purpose of this study was to assess the presence of Nav1.9 channels in medial prefrontal cortex (mPFC) layer II and V pyramidal neurons in young (20-day old), late adolescent (60-day old), and adult (6- to 7-month old) rats. First, we demonstrated that layer II and V mPFC pyramidal neurons in slices obtained from young rats exhibited a TTX-resistant, low-threshold, noninactivating, and voltage-dependent Na+ current. The mRNA expression of the SCN11a gene (which encodes the Nav1.9 channel) in mPFC tissue was significantly higher in young rats than in late adolescent and adult rats. Nav1.9 protein was immunofluorescently labeled in mPFC cells in slices and analyzed via confocal microscopy. Nav1.9 immunolabeling was present in layer II and V mPFC pyramidal neurons and was more prominent in the neurons of young rats than in the neurons of late adolescent and adult rats. We conclude that Nav1.9 channels are expressed in layer II and V mPFC pyramidal neurons and that Nav1.9 protein expression in the mPFC pyramidal neurons of late adolescent and adult rats is lower than that in the neurons of young rats. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1371-1384, 2017.
Collapse
Affiliation(s)
- Maciej Gawlak
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| | - Bartłomiej Szulczyk
- Department of Drug Technology and Pharmaceutical Biotechnology, The Medical University of Warsaw, Warsaw, Poland
| | - Adam Berłowski
- Department of Physiology and Pathophysiology, The Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Grzelka
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| | - Anna Stachurska
- Department of Molecular Biology, The Medical University of Warsaw, Warsaw, Poland
| | - Justyna Pełka
- Department of Physiology and Pathophysiology, The Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Czarzasta
- Laboratory of Experimental and Clinical Physiology, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Małecki
- Department of Molecular Biology, The Medical University of Warsaw, Warsaw, Poland
| | - Przemysław Kurowski
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| | - Ewa Nurowska
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| | - Paweł Szulczyk
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|