Thus, our results suggest that the key event leading to induction of the erythroid fate is the assembly of the GATA1/TAL1/LMO2/LDB1 complex. The role of in iEP generation is more ambiguous because its protein is not known to interact with GATA1, TAL1, or LMO2. development. Graphical Abstract Open in a separate window Introduction Although several factors are known to participate in the conserved genetic program instructing development of committed erythroid progenitors, the minimal combination of factors required for direct induction of erythroid cell fate remains unknown. The identification of the key players controlling red blood cell (RBC) development is important for understanding basic biology and can be used to study and recapitulate erythropoiesis in?vitro as well as to model and develop Nitidine chloride new therapies for RBC disorders (Tsiftsoglou et?al., 2009). Fate decisions in erythropoiesis have been investigated extensively, focusing on lineage-specific transcription factors and cofactors as the main drivers of the process (Cantor and Orkin, 2002, Shivdasani and Orkin, 1996). Genes found Nitidine chloride to be essential for normal RBC development in mice include (Mead et?al., 2001, Palis, 2014). However, the factors constituting the core transcriptional machinery that initiates and specifies erythroid cell fate are still unknown. A major obstacle for defining core transcriptional networks is the difficulty of discriminating instructive factors from permissive factors. Numerous studies have exhibited that it is possible to directly convert a mature cell type into another, bypassing the pluripotent Nitidine chloride state, using a defined set of lineage-instructive transcription Nitidine chloride factors (Jopling et?al., 2011, Takahashi, 2012). This approach, called direct lineage reprogramming, can yield a wide range of clinically relevant cell types, such as neurons, cardiomyocytes, and hepatocytes (Huang et?al., 2011, Ieda et?al., 2010, Sekiya and Suzuki, 2011, Vierbuchen et?al., 2010). Because the converted cells resemble their bona fide counterparts in terms of phenotype and function, direct lineage reprogramming is currently a widely investigated approach for generating defined cell types for regenerative medicine. In contrast to loss-of-function studies, direct reprogramming distinguishes absolutely essential cell fate-inducing factors from merely permissive factors, revealing the grasp regulators of specific cell lineages (Vierbuchen and Wernig, 2011). Therefore, we reasoned that direct lineage reprogramming is an unambiguous method for defining the core transcriptional machinery directing RBC development. Several laboratories have described methods for reprogramming differentiated somatic cells to hematopoietic progenitors with multilineage potential (Batta et?al., 2014, Pereira et?al., 2013, Riddell et?al., 2014, Szabo et?al., 2010), whereas others have reported protocols of direct induction to the erythroid lineage starting from B cells (Sadahira et?al., 2012) and pluripotent cell sources (Weng and Sheng, 2014). However, none of these studies have shown strong erythroid-restricted fate conversion from non-hematopoietic differentiated somatic cells. Here we identify the transcription factors (GTLM) as the minimal set of factors for direct conversion of mouse and human fibroblasts into erythroid progenitors. The resulting cells, which we term induced erythroid progenitors/precursors (iEPs), resemble bona fide erythroid cells in terms of morphology, colony-forming capacity, and gene expression. While murine GTLM iEPs express both embryonic and adult globin genes, the addition of or induces a switch in globin gene expression to generate iEPs with a?predominant definitive-type globin expression pattern. This approach can be used as a model for understanding, controlling, and recapitulating erythroid lineage development and disease. Results A Combination of Transcription Factors Induces the Erythroid Fate in Murine Fibroblasts We hypothesized that overexpression of transcription factors involved in hematopoietic Nitidine chloride and, specifically, erythroid development in fibroblasts could directly convert these cells into erythroid progenitors or precursors. A retroviral library was created from mouse fetal liver (FL) cDNA expressing the coding region of 63 candidate factors (Table S1). Adult tail tip fibroblasts (TTFs) were derived from erythroid lineage-tracing mice (Heinrich et?al., 2004), which express the Rabbit Polyclonal to p70 S6 Kinase beta (phospho-Ser423) yellow fluorescent protein (eYFP) from the locus in all cells that have expressed the erythropoietin receptor (locus) transcript at any stage of their development (Physique?1A). In?vivo, the expression of eYFP is first detected in bipotent progenitors of megakaryocytes and erythrocytes (pre-MegEs) and is subsequently robustly expressed in erythroid progenitors (Singbrant et?al., 2011). Importantly, eYFP was never detected in other hematopoietic lineages or cell types examined. TTF cultures were carefully depleted of hematopoietic cells by magnetic separation using a cocktail of nine hematopoietic antibodies (Experimental Procedures) and passaged at least three times prior to transduction to obtain real fibroblast cultures. The primary readout for erythroid lineage conversion was the formation of colonies of eYFP+ (EpoR+) round cells. Open in a separate window Physique?1 Forced Expression of Reprograms Murine Adult Fibroblasts into Erythroid Progenitors (A) Experimental design for transcription factor-mediated reprogramming of erythroid reporter (from the factor cocktail completely abrogated iEP formation (Physique?1C; Physique?S1). Notably, TTF reprogramming to iEPs was significantly enhanced using only.