TY - CHAP AU - Zöldi, Miklós AU - Katona, István ED - Maccarrone, Mauro TI - STORM Super-Resolution Imaging of CB1 Receptors in Tissue Preparations T2 - Endocannabinoid signaling: methods and protocols PB - Springer US CY - New York, New York SN - 9781071627303 T3 - Methods in Molecular Biology, ISSN 1064-3745 ; 2576. PY - 2023 SP - 437 EP - 451 PG - 15 DO - 10.1007/978-1-0716-2728-0_36 UR - https://m2.mtmt.hu/api/publication/33115714 ID - 33115714 AB - Single-molecule localization microscopy (SMLM) opened new possibilities to study the spatial arrangement of molecular distribution and disease-associated redistribution at a previously unprecedented resolution that was not achievable with optical microscopy approaches. Recent discoveries based on SMLM techniques uncovered specific nanoscale organizational principles of signaling proteins in several biological systems including the chemical synapses in the brain. Emerging data suggest that the spatial arrangement of the molecular players of the endocannabinoid system is also precisely regulated at the nanoscale level in synapses and in other neuronal and glial subcellular compartments. The precise nanoscale distribution pattern is likely to be important to subserve several specific signaling functions of this important messenger system in a cell-type- and subcellular domain-specific manner. LA - English DB - MTMT ER - TY - JOUR AU - Cserép, Csaba AU - Schwarcz, Dóra Anett AU - Pósfai, Balázs AU - László, Zsófia Ilona AU - Kellermayer, Anna AU - Környei, Zsuzsanna AU - Kisfali, Máté AU - Nyerges, Miklós AU - Lele, Zsolt AU - Katona, István AU - Dénes, Ádám TI - Microglial control of neuronal development via somatic purinergic junctions JF - CELL REPORTS J2 - CELL REP VL - 40 PY - 2022 IS - 12 PG - 22 SN - 2211-1247 DO - 10.1016/j.celrep.2022.111369 UR - https://m2.mtmt.hu/api/publication/33111942 ID - 33111942 N1 - “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, 1083, Hungary “Momentum” Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest, 1083, Hungary University of Dundee, School of Medicine, Dundee, DD1 9SY, United Kingdom Szentágothai János Doctoral School of Neurosciences, Semmelweis University, Budapest, 1083, Hungary Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, United States Export Date: 27 March 2023 Correspondence Address: Cserép, C.; “Momentum” Laboratory of Neuroimmunology, Hungary; email: cserep.csaba@koki.hu Correspondence Address: Adam Denes; “Momentum” Laboratory of Neuroimmunology, Hungary; email: denes.adam@koki.hu Chemicals/CAS: lipocortin 5, 111237-10-6 Funding details: Magyar Tudományos Akadémia, MTA, LP2016-4/2016, LP2022-5/2022, ÚNKP-20-3-II, ÚNKP-20-5, ÚNKP-21-4, ÚNKP-21-5 Funding details: Nemzeti Kutatási Fejlesztési és Innovációs Hivatal, NKFIH, 129961 Funding details: Richter Gedeon Talentum Alapítvány, 2017-1.2.1-NKP-2017-00002, 2020-1.2.4-TÉT-IPARI-2021-00005 Funding details: Innovációs és Technológiai Minisztérium Funding text 1: We thank Drs. László Barna and Pál Vági for kindly providing microscopy support and Dóra Gali-Györkei, Balázs Pintér, and Erika Tischler for excellent technical assistance. The study was supported by “ Momentum ” research grants from the Hungarian Academy of Sciences ( LP2016-4/2016 and LP2022-5/2022 to Á.D.), ERC-CoG 724994 (to Á.D.), the János Bolyai Research Scholarship of the Hungarian Academy of Sciences (to C.C.), ÚNKP-20-3-II and ÚNKP-21-4 (to B.P.) and ÚNKP-20-5 and ÚNKP-21-5 (to C.C.), the New National Excellence Program of the Ministry for Innovation and Technology , the Gedeon Richter's Talentum Foundation (to A.D.S.), and by 2020-1.2.4-TÉT-IPARI-2021-00005 (to Á.D.). I.K. was supported by the National Brain Research Program ( 2017-1.2.1-NKP-2017-00002 ) and by the National Research, Development and Innovation Office , Hungary (Frontier Program 129961). The funding institutions had no role in the conceptualization, design, data collection, analysis, decision to publish, or preparation of the manuscript. LA - English DB - MTMT ER - TY - JOUR AU - Prokop, Susanne Clara AU - Ábrányi-Balogh, Péter AU - Barti, Benjámin AU - Vámosi, Márton György AU - Zöldi, Miklós AU - Barna, László AU - Katona-Urbán, Gabriella AU - Tóth, András AU - Dudok, Barna AU - Egyed, Attila AU - Deng, Hui AU - Leggio, Gian Marco AU - Hunyady, László AU - van der Stelt, Mario AU - Keserű, György Miklós AU - Katona, István TI - PharmacoSTORM nanoscale pharmacology reveals cariprazine binding on Islands of Calleja granule cells JF - NATURE COMMUNICATIONS J2 - NAT COMMUN VL - 12 PY - 2021 IS - 1 PG - 19 SN - 2041-1723 DO - 10.1038/s41467-021-26757-z UR - https://m2.mtmt.hu/api/publication/32491765 ID - 32491765 N1 - Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest, Hungary School of Ph.D. Studies, Semmelweis University, Budapest, Hungary Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States Nikon Center of Excellence for Neuronal Imaging, Institute of Experimental Medicine, Budapest, Hungary Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary MTA-SE Laboratory of Molecular Physiology, Eötvös Loránd Research Network, Budapest, Hungary Department of Neurosurgery, Stanford University, Stanford, CA, United States Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University & Oncode Institute, Leiden, Netherlands Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy Cited By :3 Export Date: 18 May 2022 Correspondence Address: Katona, I.; Momentum Laboratory of Molecular Neurobiology, Hungary; email: katona@koki.hu Chemicals/CAS: cariprazine, 839712-12-8, 1083076-69-0, 955400-75-6; cariprazine; Piperazines; Receptors, Dopamine D2; Receptors, Dopamine D3 Funding details: National Institutes of Health, NIH, R01DA044925, R01NS099457 Funding details: Nederlandse Organisatie voor Wetenschappelijk Onderzoek, NWO Funding details: Ministero dell’Istruzione, dell’Università e della Ricerca, MIUR, 201779W93T Funding details: Università di Catania, UNICT Funding details: Innovációs és Technológiai Minisztérium, ÚNKP-19-3-III-SE-16, ÚNKP-20-3-II-SE-33 Funding details: National Research, Development and Innovation Office, 129961, PD124598, VEKOP-2.3.3-15-2016-00013 Funding text 1: The authors are grateful to B. Pintér and E. Tischler for their laboratory support. The authors thank Drs. J. Brunner, K. Mackie, P. Somogyi, and J. Szabadics for valuable discussions and comments on the manuscript. The help of Drs. P. Vági, C. Pongor, the Nikon Microscopy Center at the Institute of Experimental Medicine, Nikon Europe B.V., Nikon Austria GmbH, and Auro-Science Consulting is acknowledged for kindly providing microscopy support. The authors are also grateful to Drs. E. Horváth, K. Kenesei, M. Kisfali, Z. Lele, Z. László, G. Balla, F. Mógor, and J. Glavinics for their experimental advice in laboratory work. The authors are indebted to Dr. B. Gereben for providing the Grenier Luminometer and HEK 293 cells. The authors also thank the help of D. Kiss and Á. Kelemen for their help in chemical modeling and synthesis, and M. Jiang for performing the docking studies with DH-463. This study was supported by the National Brain Research Program (2017-1.2.1-NKP-2017-00002 for I.K. and G.M.K.), by the National Research, Development and Innovation Office, Hungary (VEKOP-2.3.3-15-2016-00013 for super-resolution microscopy development for I.K.; Frontier Program 129961 for cannabinoid research for I.K.; postdoctoral fellowship PD124598 for P.Á.-B., EFOP-3.6.3-VEKOP-16-2017-00009 scholarship for M.V.). I.K. holds the Naus Family Chair in Addiction Sciences in the Department of Psychological and Brain Sciences at Indiana University Bloomington and his work is also supported by the National Institutes of Health (R01NS099457 and R01DA044925). This study was also supported by the New National Excellence Program of the Ministry for Innovation and Technology (ÚNKP-20-3-II-SE-33 and ÚNKP-19-3-III-SE-16 to S.P. and B.B.); by the VICI-grant from the Netherlands Organization for Scientific Research and funding from Oncode Institute to M.v.d.S.; the Italian Ministry of University and Research (PRIN 2017-Prot. 201779W93T to G.M.L.) and the University of Catania Intramural Funds (Starting Grant 2020 to G.M.L.). LA - English DB - MTMT ER - TY - JOUR AU - Klein, Peter M. AU - Parihar, Vipan K. AU - Szabo, Gergely G. AU - Zöldi, Miklós AU - Angulo, Maria C. AU - Allen, Barrett D. AU - Amin, Amal N. AU - Nguyen, Quynh-Anh AU - Katona, István AU - Baulch, Janet E. AU - Limoli, Charles L. AU - Soltesz, Ivan TI - Detrimental impacts of mixed-ion radiation on nervous system function JF - NEUROBIOLOGY OF DISEASE J2 - NEUROBIOL DIS VL - 151 PY - 2021 SN - 0969-9961 DO - 10.1016/j.nbd.2021.105252 UR - https://m2.mtmt.hu/api/publication/31830409 ID - 31830409 N1 - Department of Neurosurgery, Stanford University, Palo Alto, CA 94305, United States Department of Radiation Oncology, University of California, Irvine, CA 92697, United States Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1083, Hungary Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, United States Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA 94305, United States Cited By :4 Export Date: 8 February 2022 CODEN: NUDIE Correspondence Address: Klein, P.M.1201 Welch Rd, United States; email: kleinp@stanford.edu Chemicals/CAS: helium, 7440-59-7; iron, 14093-02-8, 53858-86-9, 7439-89-6; oxygen, 7782-44-7; proton, 12408-02-5, 12586-59-3; silicon, 7440-21-3 Funding details: 129961 Funding details: National Aeronautics and Space Administration, NASA, NNX15AI22G Funding text 1: This work was supported by NASA Specialized Center of Research grant NNX15AI22G (CLL, IS) and by the National Research, Development and Innovation Office of Hungary Frontier Program grant 129961 (MZ, IK) . Thank you to all members of the NASA Space Radiation Laboratory for their invaluable assistance in the irradiation of animals. AB - Galactic cosmic radiation (GCR), composed of highly energetic and fully ionized atomic nuclei, produces diverse deleterious effects on the body. In researching the neurological risks of GCR exposures, including during human spaceflight, various ground-based single-ion GCR irradiation paradigms induce differential disruptions of cellular activity and overall behavior. However, it remains less clear how irradiation comprising a mix of multiple ions, more accurately recapitulating the space GCR environment, impacts the central nervous system. We therefore examined how mixed-ion GCR irradiation (two similar 5-6 beam combinations of protons, helium, oxygen, silicon and iron ions) influenced neuronal connectivity, functional generation of activity within neural circuits and cognitive behavior in mice. In electrophysiological recordings we find that space-relevant doses of mixed-ion GCR preferentially alter hippocampal inhibitory neurotransmission and produce related disruptions in the local field potentials of hippocampal oscillations. Such underlying perturbation in hippocampal network activity correspond with perturbed learning, memory and anxiety behavior. LA - English DB - MTMT ER - TY - JOUR AU - Csapóné Miczán, Vivien AU - Kelemen, Krisztina AU - Glavinics, Judit Réka AU - László, Zsófia Ilona AU - Barti, Benjámin AU - Kenesei, Kata AU - Kisfali, Máté AU - Katona, István TI - NECAB1 and NECAB2 are Prevalent Calcium-Binding Proteins of CB1/CCK-Positive GABAergic Interneurons JF - CEREBRAL CORTEX J2 - CEREB CORTEX VL - 31 PY - 2021 IS - 3 SP - 1786 EP - 1806 PG - 21 SN - 1047-3211 DO - 10.1093/cercor/bhaa326 UR - https://m2.mtmt.hu/api/publication/31659826 ID - 31659826 N1 - Funding Agency and Grant Number: New National Excellence Program of the Ministry of Human Capacities [UNKP-18-3]; National Brain Research Program [2017-1.2.1-NKP-2017-00002]; National Research, Development and Innovation Office, Hungary [129961, KH124972]; National Institutes of Health [R01NS099457, R01DA044925] Funding text: New National Excellence Program of the Ministry of Human Capacities (UNKP-18-3); National Brain Research Program (2017-1.2.1-NKP-2017-00002); National Research, Development and Innovation Office, Hungary (Frontier Program 129961, KH124972); National Institutes of Health (R01NS099457, R01DA044925). I.K. also holds the Naus Family Chair in Addiction Sciences in the Department of Psychological and Brain Sciences at Indiana University Bloomington. LA - English DB - MTMT ER - TY - JOUR AU - László, Zsófia Ilona AU - Lele, Zsolt AU - Zöldi, Miklós AU - Csapóné Miczán, Vivien AU - Mógor, Fruzsina AU - Simon, Gabriel M. AU - Mackie, Ken AU - Kacskovics, Imre AU - Cravatt, Benjamin F. AU - Katona, István TI - ABHD4-dependent developmental anoikis safeguards the embryonic brain JF - NATURE COMMUNICATIONS J2 - NAT COMMUN VL - 11 PY - 2020 IS - 1 PG - 16 SN - 2041-1723 DO - 10.1038/s41467-020-18175-4 UR - https://m2.mtmt.hu/api/publication/31523670 ID - 31523670 N1 - Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, 1450 Budapest Pf. 67., Budapest, Hungary School of Ph.D. Studies, Semmelweis University, Budapest, Hungary Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary The Skaggs Institute for Chemical Biology, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92307, United States Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, United States Department of Immunology, Eötvös Loránd University, Pázmány Péter stny 1/A., Budapest, 1117, Hungary ImmunoGenes Ltd, Makkosi út 86., Budakeszi, 2092, Hungary Cited By :11 Export Date: 4 May 2023 Correspondence Address: Katona, I.; Momentum Laboratory of Molecular Neurobiology, 1450 Budapest Pf. 67., Hungary; email: katona@koki.hu Chemicals/CAS: protein, 67254-75-5; lysophospholipase, 9001-85-8; ABHD4 protein, human; Lysophospholipase Funding details: 129961, K116915, VEKOP-2.3.3-15-2016-00013, VKSZ-14-1-2O15-0155 Funding details: National Institutes of Health, NIH, DA011322, DA037660, R01DA044925 Funding details: National Institute on Drug Abuse, NIDA, K05DA021696 Funding details: National Institute of Neurological Disorders and Stroke, NINDS, R01NS099457 Funding details: Semmelweis Egyetem, EFOP-3.6.3-VEKOP-16-2017-00009 Funding text 1: The authors thank Dr. E. Horváth, B. Pintér, E. Tischler for laboratory support, Drs. G. Balogh, Z. Balogi, A. Dorning, B. Dudok, M. Melis, M. Péter, S. Prokop, L. Vígh for help with preliminary experiments and comments. The authors appreciate the help of Dr. Ozge Gunduz-Cinar and Dr. Andrew Holmes with the RNAscope protocol. The authors are grateful to the IEM Medical Genetics Unit for mouse colony management, to S.S.J. Hu, K. Nagy for antibody generation, to Dr. B. Gereben for providing HEK293 cells and to K. Balogh for artwork. The help of Dr. L. Barna, the Nikon Microscopy Center at the Institute of Experimental Medicine, Nikon Europe B.V., Nikon Austria GmbH and Auro-Science Consulting is acknowledged for kindly providing microscopy support. This work was supported by the National Brain Research Program (2017-1.2.1-NKP-2017-00002), by the National Research, Development and Innovation Office, Hungary (VKSZ-14-1-2O15-0155 for antibody generation; VEKOP-2.3.3-15-2016-00013 for super-resolution microscopy development; K116915 for ABHD4 research and Frontier Program 129961 for cannabinoid research). I.K. holds the Naus Family Chair in Addiction Sciences in the Department of Psychological and Brain Sciences at Indiana University Bloomington and his work is also supported by the National Institutes of Health (R01NS099457 and R01DA044925). Additional support was provided by the Semmelweis University Grant (EFOP-3.6.3-VEKOP-16-2017-00009 to Z.I.L.), the US National Institutes of Health Grant (DA021696 and DA011322) to K.M. and (DA037660) to B.F.C. LA - English DB - MTMT ER - TY - JOUR AU - Cserép, Csaba AU - Pósfai, Balázs AU - Lénárt, Nikolett AU - Fekete, Rebeka AU - László, Zsófia Ilona AU - Lele, Zsolt AU - Orsolits, Barbara AU - Molnár, Gábor AU - Heindl, S AU - Schwarcz, Dóra Anett AU - Ujvári, Katinka AU - Környei, Zsuzsanna AU - Tóth, Krisztina AU - Cserépné Szabadits, Eszter AU - Sperlágh, Beáta AU - Baranyi, Mária AU - Csiba, László AU - Hortobágyi, Tibor AU - Maglóczky, Zsófia AU - Martinecz, Bernadett AU - Szabó, Gábor AU - Erdélyi, Ferenc AU - Szipőcs, Róbert AU - Tamkun, MM AU - Gesierich, B AU - Duering, M AU - Katona, István AU - Liesz, A AU - Tamás, Gábor AU - Dénes, Ádám TI - Microglia monitor and protect neuronal function through specialized somatic purinergic junctions JF - SCIENCE J2 - SCIENCE VL - 367 PY - 2020 IS - 6477 SP - 528 EP - 537 PG - 11 SN - 0036-8075 DO - 10.1126/science.aax6752 UR - https://m2.mtmt.hu/api/publication/31294298 ID - 31294298 LA - English DB - MTMT ER - TY - JOUR AU - Bernal-Chico, Ana AU - Manterola, Andrea AU - Cipriani, Raffaela AU - Katona, István AU - Matute, Carlos AU - Mato, Susana TI - P2x7 receptors control demyelination and inflammation in the cuprizone model JF - BRAIN BEHAVIOR IMMUNITY - HEALTH J2 - BRAIN BEHAVIOR IMMUNITY - HEALTH VL - 4 PY - 2020 PG - 14 SN - 2666-3546 DO - 10.1016/j.bbih.2020.100062 UR - https://m2.mtmt.hu/api/publication/31289763 ID - 31289763 N1 - Department of Neurosciences, School of Medicine, University of the Basque Country UPV/EHU, Leioa, 48940, Spain Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, 28031, Spain Biocruces, Barakaldo, Bizkaia 48903, Spain Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1083, Hungary Cited By :10 Export Date: 8 June 2023 Correspondence Address: Mato, S.; Department of Neurosciences, Spain; email: susana.mato@ehu.eus Correspondence Address: Matute, C.; Department of Neurosciences, Spain; email: carlos.matute@ehu.eus Chemicals/CAS: brilliant blue g, 6104-58-1; cuprizone, 370-81-0 Tradenames: jnj 47965567, Janssen Research and Development Manufacturers: Biorad; Janssen Research and Development AB - The contribution of P2x7 receptors to multiple sclerosis remains controversial, as both detrimental and beneficial effects resulting from P2x7 receptor loss-of-function have been reported in autoimmune models of the disease. Here we investigated the relevance of P2x7 receptors to de- and remyelination in the cuprizone model of T cell-independent myelin degeneration. Primary demyelination was induced by administration of 0.3% cuprizone in the diet for 3 and 6 weeks. Remyelination was studied in mice treated with the P2x7 receptor antagonists Brilliant Blue G (BBG, 50 mg/Kg) and JNJ-47965567 (30 mg/Kg) for 2 weeks following 6 weeks of cuprizone challenge. Toxic demyelination induced a robust up-regulation of P2x7 receptors mainly localized on microglial cells. In parallel, we measured increased expression of several NLPR3-inflammasome and M1 polarization-associated genes in demyelinated tissue. Notably, mice deficient in P2x7 receptors exhibited attenuated demyelination, reduced presence of M1 microglia and reactive astrocytes as well as blunted expression of pro-inflammatory genes in response to cuprizone feeding. Nevertheless, blockade of P2x7 receptors during the remyelination phase did not improve the extent of myelin recovery nor attenuated glial reaction and inflammation in damaged white matter. These findings suggest that P2x7 receptors drive T cell-independent inflammation and demyelination, but are not relevant to regenerative responses involved in myelin repair. LA - English DB - MTMT ER - TY - JOUR AU - Katona, István TI - A Molecular Collapse and the Mental “Falling Down” JF - NEURON J2 - NEURON VL - 105 PY - 2020 IS - 6 SP - 956 EP - 958 PG - 3 SN - 0896-6273 DO - 10.1016/j.neuron.2020.02.026 UR - https://m2.mtmt.hu/api/publication/31254787 ID - 31254787 N1 - Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, United States Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest, 1083, Hungary Export Date: 4 May 2023 CODEN: NERNE Correspondence Address: Katona, I.; Department of Psychological and Brain Sciences, United States; email: katona@koki.hu Chemicals/CAS: Endocannabinoids LA - English DB - MTMT ER - TY - JOUR AU - László, Zsófia Ilona AU - Bercsényi, Kinga AU - Mayer, Mátyás AU - Lefkovics, Kornél AU - Szabó, Gábor AU - Katona, István AU - Lele, Zsolt TI - N-cadherin (Cdh2) Maintains Migration and Postmitotic Survival of Cortical Interneuron Precursors in a Cell-Type-Specific Manner JF - CEREBRAL CORTEX J2 - CEREB CORTEX VL - 30 PY - 2020 IS - 3 SP - 1318 EP - 1329 PG - 12 SN - 1047-3211 DO - 10.1093/cercor/bhz168 UR - https://m2.mtmt.hu/api/publication/30866046 ID - 30866046 N1 - Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony utca 43, Budapest, H-1083, Hungary Laboratory of Molecular Biology and Genetics, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest, Hungary Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom Cited By :8 Export Date: 5 March 2024 CODEN: CECOE Correspondence Address: Lele, Z.; Momentum Laboratory of Molecular Neurobiology, Szigony utca 43, Hungary; email: lele.zsolt@koki.mta.hu Chemicals/CAS: Cadherins; Cdh2 protein, mouse Funding details: 2017-1.2.1-NKP-2017-00002, K 116915 Funding details: SH/7/2/18 Funding details: Wellcome Trust, WT, 090946/Z/09/Z Funding details: European Research Council, ERC, 243153 Funding details: Semmelweis Egyetem, EFOP-3.6.3-VEKOP-16-2017-00009 Funding details: Hungarian Scientific Research Fund, OTKA, F 68177 Funding details: Magyar Tudományos Akadémia, MTA, LP2013-54/2013 Funding details: National Research, Development and Innovation Office, VKSZ_14-1-2O15-0155 Funding text 1: Hungarian Scientific Research Fund (OTKA, F 68177 to Z.L.); the European Research Council Grant (243153 to I.K.); the National Research, Development and Innovation Office, Hungary (VKSZ_14-1-2O15-0155 to I.K.); the Hungarian Brain Research Program (2017-1.2.1-NKP-2017-00002 to I.K.); the Hungarian Scientific Research Fund (OTKA, K 116915 to I.K.); the Momentum Program of the Hungarian Academy of Sciences (LP2013-54/2013 to I.K.); the Wellcome Trust International Senior Research Fellowship (090946/Z/09/Z to I.K.); Semmelweis University Predoctoral Grant (EFOP-3.6.3-VEKOP-16-2017-00009 to Z.I.L.); the Swiss Contribution grant (SH/7/2/18 to I.K.). AB - The multiplex role of cadherin-based adhesion complexes during development of pallial excitatory neurons has been thoroughly characterized. In contrast, much less is known about their function during interneuron development. Here, we report that conditional removal of N-cadherin (Cdh2) from postmitotic neuroblasts of the subpallium results in a decreased number of Gad65-GFP-positive interneurons in the adult cortex. We also found that interneuron precursor migration into the pallium was already delayed at E14. Using immunohistochemistry and TUNEL assay in the embryonic subpallium, we excluded decreased mitosis and elevated cell death as possible sources of this defect. Moreover, by analyzing the interneuron composition of the adult somatosensory cortex, we uncovered an unexpected interneuron-type-specific defect caused by Cdh2-loss. This was not due to a fate-switch between interneuron populations or altered target selection during migration. Instead, potentially due to the migration delay, part of the precursors failed to enter the cortical plate and consequently got eliminated at early postnatal stages. In summary, our results indicate that Cdh2-mediated interactions are necessary for migration and survival during the postmitotic phase of interneuron development. Furthermore, we also propose that unlike in pallial glutamatergic cells, Cdh2 is not universal, rather a cell type-specific factor during this process. LA - English DB - MTMT ER -