Vis enkel innførsel

dc.contributor.authorSepponen, Kirsi
dc.contributor.authorLundin, Karolina
dc.contributor.authorYohannes, Dawit A.
dc.contributor.authorVuoristo, Sanna
dc.contributor.authorBalboa, Diego
dc.contributor.authorPoutanen, Matti
dc.contributor.authorOhlsson, Claes
dc.contributor.authorHustad, Steinar
dc.contributor.authorBifulco, Ersilia
dc.contributor.authorPaloviita, Pauliina
dc.contributor.authorOtonkoski, Timo
dc.contributor.authorRitvos, Olli
dc.contributor.authorSainio, Kirsi
dc.contributor.authorTapanainen, Juha S.
dc.contributor.authorTuuri, Timo
dc.date.accessioned2023-01-18T10:27:33Z
dc.date.available2023-01-18T10:27:33Z
dc.date.created2022-10-22T12:26:26Z
dc.date.issued2022
dc.identifier.issn0301-4681
dc.identifier.urihttps://hdl.handle.net/11250/3044232
dc.description.abstractNuclear receptor subfamily 5 group A member 1 (NR5A1) encodes steroidogenic factor 1 (SF1), a key regulatory factor that determines gonadal development and coordinates endocrine functions. Here, we have established a stem cell-based model of human gonadal development and applied it to evaluate the effects of NR5A1 during the transition from bipotential gonad to testicular cells. We combined directed differentiation of human induced pluripotent stem cells (46,XY) with activation of endogenous NR5A1 expression by conditionally-inducible CRISPR activation. The resulting male gonadal-like cells expressed several Sertoli cell transcripts, secreted anti-Müllerian hormone and responded to follicle-stimulating hormone by producing sex steroid intermediates. These characteristics were not induced without NR5A1 activation. A total of 2691 differentially expressed genetic elements, including both coding and non-coding RNAs, were detected immediately following activation of NR5A1 expression. Of those, we identified novel gonad-related putative NR5A1 targets, such as SCARA5, which we validated also by immunocytochemistry. In addition, NR5A1 activation was associated with dynamic expression of multiple gonad- and infertility-related differentially expressed genes. In conclusion, by combining targeted differentiation and endogenous activation of NR5A1 we have for the first time, been able to examine in detail the effects of NR5A1 in early human gonadal cells. The model and results obtained provide a useful resource for future investigations exploring the causative reasons for gonadal dysgenesis and infertility in humans.en_US
dc.language.isoengen_US
dc.publisherElsevieren_US
dc.rightsNavngivelse 4.0 Internasjonal*
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/deed.no*
dc.titleSteroidogenic factor 1 (NR5A1) induces multiple transcriptional changes during differentiation of human gonadal-like cellsen_US
dc.typeJournal articleen_US
dc.typePeer revieweden_US
dc.description.versionpublishedVersionen_US
dc.rights.holderCopyright 2022 the authorsen_US
cristin.ispublishedtrue
cristin.fulltextoriginal
cristin.qualitycode1
dc.identifier.doi10.1016/j.diff.2022.08.001
dc.identifier.cristin2063944
dc.source.journalDifferentiationen_US
dc.source.pagenumber83-100en_US
dc.identifier.citationDifferentiation. 2022, 128, 83-100.en_US
dc.source.volume128en_US


Tilhørende fil(er)

Thumbnail

Denne innførselen finnes i følgende samling(er)

Vis enkel innførsel

Navngivelse 4.0 Internasjonal
Med mindre annet er angitt, så er denne innførselen lisensiert som Navngivelse 4.0 Internasjonal