RT Journal Article SR Electronic T1 Dissecting transcriptomic signatures of neuronal differentiation and maturation using iPSCs JF bioRxiv FD Cold Spring Harbor Laboratory SP 380758 DO 10.1101/380758 A1 EE Burke A1 JG Chenoweth A1 JH Shin A1 L Collado-Torres A1 SK Kim A1 N Micali A1 Y Wang A1 RE Straub A1 DJ Hoeppner A1 HY Chen A1 A Lescure A1 K Shibbani A1 GR Hamersky A1 BN Phan A1 WS Ulrich A1 C Valencia A1 A Jaishankar A1 AJ Price A1 A Rajpurohit A1 SA Semick A1 R Bürli A1 JC Barrow A1 DJ Hiler A1 SC Page A1 K Martinowich A1 TM Hyde A1 JE Kleinman A1 KF Berman A1 JA Apud A1 AJ Cross A1 NJ Brandon A1 DR Weinberger A1 BJ Maher A1 RDG McKay A1 AE Jaffe YR 2018 UL http://biorxiv.org/content/early/2018/07/31/380758.abstract AB Human induced pluripotent stem cells (hiPSCs) are a powerful model of neural differentiation and maturation. We present a hiPSC transcriptomics resource on corticogenesis from 5 iPSC donor and 13 subclonal lines across nine time points over 5 broad conditions: self-renewal, early neuronal differentiation, neural precursor cells (NPCs), assembled rosettes, and differentiated neuronal cells that were validated using electrophysiology. We identified widespread changes in the expression of individual transcript features and their splice variants, gene networks, and global patterns of transcription. We next demonstrated that co-culturing human NPCs with rodent astrocytes resulted in mutually synergistic maturation, and that cell type-specific expression data can be extracted using only sequencing read alignments without potentially disruptive cell sorting. We lastly developed and validated a computational tool to estimate the relative neuronal maturity of iPSC-derived neuronal cultures and human brain tissue, which were maturationally heterogeneous but contained subsets of cells most akin to adult human neurons.