Categories
ETA Receptors

To ensure the quality of starting iPSCs, the cells were thawed in L7? hPSC medium and L7? Matrix, serially subcultured using L7? hPSC passaging solution and underwent standard characterization and safety studies

To ensure the quality of starting iPSCs, the cells were thawed in L7? hPSC medium and L7? Matrix, serially subcultured using L7? hPSC passaging solution and underwent standard characterization and safety studies. S4: Comparison of cell number and viability after harvest. Human-umHHinduced pluripotent stem cells were harvested on day 14 of cardiomyocyte differentiation using Liberase/TrypLE enzyme mix. Cell count and viability was measured. (A) The viable cell yield from one well of a 6-well plate was between 2.5 and 3.5??106 cells. (B) The viability of over 82% was achieved in all three runs. n.s: not significant. image_4.tif (1.4M) GUID:?158A19B6-99AE-4FAB-BBAF-49C630F9DB38 Video S1: LiPSC ER2.2-derived cardiomyocytes beatings on day 8 post differentiation. video_1.mp4 (1.7M) GUID:?BBFDD8EC-1FAF-4919-BCFB-1432965CCBE4 Video S2: LiPSC ER2.2-derived cardiomyocytes beatings on day 14 post differentiation. video_2.mp4 (2.5M) GUID:?1E779662-DE8A-495E-BD08-D1E2D5B144B0 Video S3: LiPSC 18R-derived cardiomyocytes beatings on day 11 post differentiation. video_3.mp4 (1.8M) GUID:?AE19C1D9-E8B8-4EBB-895C-12FFB1836744 Video S4: LiPSC 18R-derived cardiomyocytes beatings on day 14 post differentiation. video_4.mp4 (2.4M) GUID:?F33606CB-ACD6-493B-A481-964545EFB654 Video S5: LiPSC 18R-derived cardiomyocytes beatings on day 14 post differentiation (2 M of CHIR99021). video_5.mp4 (18M) GUID:?7DD8047A-0850-40DF-98A2-AA060C2B2A3D Video S6: LiPSC 18R-derived cardiomyocytes beatings on TCS 5861528 day 14 post differentiation (4 M of CHIR99021). video_6.mp4 (13M) GUID:?7C10930D-DBAF-4D14-BDE7-D1ED17581729 Abstract The discovery of reprogramming and generation of human-induced pluripotent stem cells (iPSCs) has revolutionized the field of regenerative medicine and opened new opportunities in cell replacement therapies. While generation of iPSCs represents a significant breakthrough, the clinical relevance of iPSCs for cell-based therapies requires generation of high-quality specialized cells through robust and reproducible directed differentiation protocols. We have recently reported manufacturing of human iPSC master cell banks (MCB) under current good manufacturing practices (cGMPs). Here, we describe the clinical potential of human iPSCs generated using this cGMP-compliant process by differentiating them into the cells from all three embryonic germ layers including ectoderm, endoderm, and mesoderm. Most importantly, we have shown that our iPSC manufacturing process and cell culture system is not biased toward a specific lineage. Following controlled induction into a specific differentiation lineage, specialized cells with morphological and cellular characteristics of neural stem cells, definitive endoderm, and cardiomyocytes were developed. We believe that these cGMP-compliant iPSCs have the potential to make various clinically relevant products suitable for cell therapy applications. and their inherent potential to differentiate into any cell type in the body, making them a precious source for clinical purposes (4). On the other hand, the increasing incidence of degenerative disorders, inefficiency of existing treatments, and the scarcity of functional primary human somatic cells are significantly increasing the demand for stem cell-based therapeutic approaches. Patient-derived iPSCs have been used to model several human genetic diseases and TCS 5861528 to successfully produce clinically relevant differentiated cells that display disease pathogenesis (5C8). Furthermore, recent progresses in the development of directed differentiation protocols using human iPSCs into various cell types (9C11) have already resulted in the start of early autologous clinical trials (12). However, establishment of a robust directed differentiated procedure starting from high-quality cells manufactured using a robust and current good manufacturing practice (cGMP)-compliant process still remain a major challenge in enabling clinical utility of iPSC-based therapies. In particular, inherent difficulties in achieving high-quality cGMP grade PSCs and their progenies is a major obstacle in cell-based therapy and should be overcome before these cell types can be used TCS 5861528 to treat diseases (13). We have recently reported TCS 5861528 the development of a cGMP-compliant process for manufacturing of human iPSCs (13) and suggested a comprehensive characterization approach (14) as an important step to develop high-quality iPSCs as input material. These iPSCs can be used at different manufacturing processes and, given their immortal status, can be utilized for many years or even decades. To demonstrate clinical relevance of these cells, we demonstrate here that our fully characterized human iPSC lines generated using cGMP-compliant process can readily differentiate into specialized cells from all three embryonic lineages with morphological and cellular characteristics of cardiomyocytes, definitive endoderm (DE), and neural stem cells (NSCs). Importantly, we also demonstrate how directed differentiation process can be further optimized to establish a robust and reproducible process as the main step in the development of a cGMP-compliant manufacturing possess to make clinical quantities of cell therapy products starting from the same iPSC lines. Materials and Methods The human iPSC lines TCS 5861528 LiPSC-ER2.2 and LiPSC-18R were generated as described before (13) under cGMP-compliant environment and were continuously maintained in feeder-independent conditions using L7? hPSCs Medium on defined L7 hPSC Matrix Mouse monoclonal to APOA4 (Lonza, FP-5020). The L7 hPSC medium included L7 hPSC basal medium (Sartorious, 04-1191F) and L7 hPSC medium supplement (Sartorious, 04-1192J). The cells were serially passaged using L7 hPSC Passaging Solution.