522 134

Full metadata record

DC FieldValueLanguage
dc.contributor.author박장환-
dc.date.accessioned2022-03-30T01:17:46Z-
dc.date.available2022-03-30T01:17:46Z-
dc.date.issued2020-07-
dc.identifier.citationEXPERIMENTAL NEUROBIOLOGY, v. 29, no. 3, page. 189-206en_US
dc.identifier.issn1226-2560-
dc.identifier.issn2093-8144-
dc.identifier.urihttps://www.en-journal.org/journal/view.html?doi=10.5607/en20021-
dc.identifier.urihttps://repository.hanyang.ac.kr/handle/20.500.11754/169526-
dc.description.abstractNeurogenic differentiation 1 (NeuroD1) is a class B basic helix-loop-helix (bHLH) transcription factor and regulates differentiation and survival of neuronal and endocrine cells by means of several protein kinases, including extracellular signal-regulated kinase (ERK). However, the effect of phosphorylation on the functions of NeuroD1 by ERK has sparked controversy based on context-dependent differences across diverse species and cell types. Here, we evidenced that ERK-dependent phosphorylation controlled the stability of NeuroD1 and consequently, regulated proneural activity in neuronal cells. A mutation at the ERK-dependent phosphorylation site, S274A, increased the half-life of NeuroD1 by blocking its ubiquitin-dependent proteasomal degradation. The S274A mutation did not interfere with either the nuclear translocation of NeuroD1 or its heterodimerization with E47, its ubiquitous partner and class A bHLH transcription factor. However, the S274A mutant increased transactivation of the E-box-mediated gene and neurite outgrowth in F11 neuroblastoma cells, compared to the wild-type NeuroD1.Transcriptome and Gene Ontology enrichment analyses indicated that genes involved in axonogenesis and dendrite development were downregulated in NeuroD1 knockout (KO) mice. Overexpression of the S274A mutant salvaged neurite outgrowth in NeuroD1-deficient mice, whereas neurite outgrowth was minimal with S274D, a phosphomimicking mutant. Our data indicated that a longer protein half-life enhanced the overall activity of NeuroD1 in stimulating downstream genes and neuronal differentiation. We propose that blocking ubiquitin-dependent proteasomal degradation may serve as a strategy to promote neuronal activity by stimulating the expression of neuron-specific genes in differentiating neurons.en_US
dc.description.sponsorshipThis study was supported by grants from the National Research Foundation of Korea (NRF-2018M3A9G1082594-to HS-K.) and the INNOPOLIS Foundation of Jeonbuk (2019-JBRD-0048-01101). We would like to thank Dr. Jacqueline E. Lee (Dep. Molecular, Cellular & Developmental Biology, University of Colorado, Boulder, USA) and Dr. Kang Ho Chul (Dep. Physiol. Ajou University, Suwon, Korea) for providing Myc-tagged NeuroD1 and HA-tagged Ubiquitin constructs, respectively.en_US
dc.language.isoenen_US
dc.publisherKOREAN SOC BRAIN & NEURAL SCIENCEen_US
dc.subjectNeurogenic differentiation factor 1en_US
dc.subjectNeurite outgrowthen_US
dc.subjectExtracellular signal-regulated kinaseen_US
dc.subjectPhosphorylationen_US
dc.titleERK Regulates NeuroD1-mediated Neurite Outgrowth via Proteasomal Degradationen_US
dc.typeArticleen_US
dc.relation.no3-
dc.relation.volume29-
dc.identifier.doi10.5607/en20021-
dc.relation.page189-206-
dc.relation.journalEXPERIMENTAL NEUROBIOLOGY-
dc.contributor.googleauthorLee, Tae-young-
dc.contributor.googleauthorCho, In-Su-
dc.contributor.googleauthorBashyal, Narayan-
dc.contributor.googleauthorNaya, Francisco J.-
dc.contributor.googleauthorTsai, Ming-Jer-
dc.contributor.googleauthorYoon, Jeong Seon-
dc.contributor.googleauthorChoi, Jung-Mi-
dc.contributor.googleauthorPark, Chang-Hwan-
dc.contributor.googleauthorKim, Sung-Soo-
dc.contributor.googleauthorSuh-Kim, Haeyoung-
dc.relation.code2020048515-
dc.sector.campusS-
dc.sector.daehakGRADUATE SCHOOL OF BIOMEDICAL SCIENCE AND ENGINEERING[S]-
dc.sector.departmentDEPARTMENT OF BIOMEDICAL SCIENCE-
dc.identifier.pidchshpark-


qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.

BROWSE