{"citation":{"mla":"Mertens, Jerome, et al. “Directly Reprogrammed Human Neurons Retain Aging-Associated Transcriptomic Signatures and Reveal Age-Related Nucleocytoplasmic Defects.” Cell Stem Cell, vol. 17, no. 6, Elsevier, 2015, pp. 705–18, doi:10.1016/j.stem.2015.09.001.","apa":"Mertens, J., Paquola, A. C. M., Ku, M., Hatch, E., Böhnke, L., Ladjevardi, S., … Gage, F. H. (2015). Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects. Cell Stem Cell. Elsevier. https://doi.org/10.1016/j.stem.2015.09.001","ieee":"J. Mertens et al., “Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects,” Cell Stem Cell, vol. 17, no. 6. Elsevier, pp. 705–718, 2015.","ama":"Mertens J, Paquola ACM, Ku M, et al. Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects. Cell Stem Cell. 2015;17(6):705-718. doi:10.1016/j.stem.2015.09.001","short":"J. Mertens, A.C.M. Paquola, M. Ku, E. Hatch, L. Böhnke, S. Ladjevardi, S. McGrath, B. Campbell, H. Lee, J.R. Herdy, J.T. Gonçalves, T. Toda, Y. Kim, J. Winkler, J. Yao, M. Hetzer, F.H. Gage, Cell Stem Cell 17 (2015) 705–718.","ista":"Mertens J, Paquola ACM, Ku M, Hatch E, Böhnke L, Ladjevardi S, McGrath S, Campbell B, Lee H, Herdy JR, Gonçalves JT, Toda T, Kim Y, Winkler J, Yao J, Hetzer M, Gage FH. 2015. Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects. Cell Stem Cell. 17(6), 705–718.","chicago":"Mertens, Jerome, Apuã C.M. Paquola, Manching Ku, Emily Hatch, Lena Böhnke, Shauheen Ladjevardi, Sean McGrath, et al. “Directly Reprogrammed Human Neurons Retain Aging-Associated Transcriptomic Signatures and Reveal Age-Related Nucleocytoplasmic Defects.” Cell Stem Cell. Elsevier, 2015. https://doi.org/10.1016/j.stem.2015.09.001."},"issue":"6","author":[{"first_name":"Jerome","full_name":"Mertens, Jerome","last_name":"Mertens"},{"full_name":"Paquola, Apuã C.M.","last_name":"Paquola","first_name":"Apuã C.M."},{"first_name":"Manching","last_name":"Ku","full_name":"Ku, Manching"},{"last_name":"Hatch","full_name":"Hatch, Emily","first_name":"Emily"},{"full_name":"Böhnke, Lena","last_name":"Böhnke","first_name":"Lena"},{"first_name":"Shauheen","last_name":"Ladjevardi","full_name":"Ladjevardi, Shauheen"},{"last_name":"McGrath","full_name":"McGrath, Sean","first_name":"Sean"},{"full_name":"Campbell, Benjamin","last_name":"Campbell","first_name":"Benjamin"},{"last_name":"Lee","full_name":"Lee, Hyungjun","first_name":"Hyungjun"},{"first_name":"Joseph R.","full_name":"Herdy, Joseph R.","last_name":"Herdy"},{"first_name":"J. Tiago","last_name":"Gonçalves","full_name":"Gonçalves, J. Tiago"},{"first_name":"Tomohisa","full_name":"Toda, Tomohisa","last_name":"Toda"},{"last_name":"Kim","full_name":"Kim, Yongsung","first_name":"Yongsung"},{"first_name":"Jürgen","full_name":"Winkler, Jürgen","last_name":"Winkler"},{"last_name":"Yao","full_name":"Yao, Jun","first_name":"Jun"},{"first_name":"Martin W","last_name":"HETZER","orcid":"0000-0002-2111-992X","id":"86c0d31b-b4eb-11ec-ac5a-eae7b2e135ed","full_name":"HETZER, Martin W"},{"last_name":"Gage","full_name":"Gage, Fred H.","first_name":"Fred H."}],"date_created":"2022-04-07T07:49:51Z","scopus_import":"1","intvolume":" 17","title":"Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects","day":"03","main_file_link":[{"url":"https://doi.org/10.1016/j.stem.2015.09.001","open_access":"1"}],"abstract":[{"lang":"eng","text":"Aging is a major risk factor for many human diseases, and in vitro generation of human neurons is an attractive approach for modeling aging-related brain disorders. However, modeling aging in differentiated human neurons has proved challenging. We generated neurons from human donors across a broad range of ages, either by iPSC-based reprogramming and differentiation or by direct conversion into induced neurons (iNs). While iPSCs and derived neurons did not retain aging-associated gene signatures, iNs displayed age-specific transcriptional profiles and revealed age-associated decreases in the nuclear transport receptor RanBP17. We detected an age-dependent loss of nucleocytoplasmic compartmentalization (NCC) in donor fibroblasts and corresponding iNs and found that reduced RanBP17 impaired NCC in young cells, while iPSC rejuvenation restored NCC in aged cells. These results show that iNs retain important aging-related signatures, thus allowing modeling of the aging process in vitro, and they identify impaired NCC as an important factor in human aging."}],"doi":"10.1016/j.stem.2015.09.001","publication_identifier":{"issn":["1934-5909"]},"month":"12","keyword":["Cell Biology","Genetics","Molecular Medicine"],"publication":"Cell Stem Cell","volume":17,"oa_version":"Published Version","status":"public","_id":"11079","pmid":1,"external_id":{"pmid":["26456686"]},"date_published":"2015-12-03T00:00:00Z","user_id":"72615eeb-f1f3-11ec-aa25-d4573ddc34fd","article_type":"original","oa":1,"quality_controlled":"1","language":[{"iso":"eng"}],"date_updated":"2022-07-18T08:44:21Z","publication_status":"published","extern":"1","publisher":"Elsevier","page":"705-718","type":"journal_article","article_processing_charge":"No","year":"2015"}