Abstract Hematopoietic differentiation of human pluripotent stem cells (hPSCs) requires orchestration of dynamic cell and gene regulatory networks but often generates blood cells that lack natural function. Here, we performed extensive single-cell transcriptomic analyses to map fate choices and gene expression patterns during hematopoietic differentiation of hPSCs and showed that oxidative metabolism was dysregulated during in vitro directed differentiation. Applying hypoxic conditions at the stage of endothelial-to-hematopoietic transition in vitro effectively promoted the development of arterial specification programs that governed the generation of hematopoietic progenitor cells (HPCs) with functional T cell potential. Following engineered expression of the anti-CD19 chimeric antigen receptor, the T cells generated from arterial endothelium-primed HPCs inhibited tumor growth both in vitro and in vivo. Collectively, our study provides benchmark datasets as a resource to further understand the origins of human hematopoiesis and represents an advance in guiding in vitro generation of functional T cells for clinical applications. INTRODUCTION Human pluripotent stem cells (hPSCs) offer a powerful resource to create in vitro models of human hematopoietic development and blood disease and have great potential for clinical cell replacement therapies. Although there are several reports of hematopoietic cells with phenotypes of hematopoietic stem cells (HSCs) or terminally differentiated hematopoietic cells generated from hPSCs ([52]1, [53]2), a robust method for the generation of these cells with consistent function has not yet been established. Thus, identifying key cellular and molecular programs required for hematopoietic specification in vitro is essential to overcome the current roadblocks. During embryonic development, hematopoietic progenitor cells (HPCs) arise from transient endothelial-like precursors known as hemogenic endothelium (HE), which are found initially in the yolk sac vascular plexus and, later, in various intraembryonic and extraembryonic vessels ([54]3, [55]4). The HE represents a distinct RUNX1-expressing subset of vascular endothelial cells (ECs) with the ability to undergo endothelial-to-hematopoietic transition (EHT) ([56]4). RUNX1 has been shown to activate the hematopoietic specification program and mediate the up-regulation of transcription factors (TFs) (e.g., GFI1 and GFI1B), which, in turn, repress endothelial gene expression ([57]5). In addition to RUNX1, many other factors have been found to regulate EHT, including TFs (e.g., GATA2 and MEIS2) and cell signaling pathways [e.g., the transforming growth factor–β (TGF-β) and Notch pathways] ([58]6–[59]9). More recently, the N-glycome and cell cycle regulation have been identified as drivers of EHT ([60]10, [61]11). Despite these discoveries, the mechanisms controlling EHT remain to be fully elucidated. A comprehensive understanding of these mechanisms is of critical importance in establishing strategies for guiding functional hematopoietic cell generation in vitro, especially in humans where the feasibility of EHT studies is limited by obvious ethical issues. Hematopoietic development is induced by signals from the microenvironment provided by anatomically distinct niches and tissue-specific vascular niches located in several hematopoietic sites, including the aorta-gonad-mesonephros (AGM), the fetal liver, and the placenta ([62]3, [63]12). In the AGM region of the mouse embryo, a variety of cellular niches—including ECs, mesenchymal cells (Mes), and mesonephros cells—have been shown to play distinct roles in HSC generation ([64]13–[65]15). In a recent study, single-cell RNA sequencing (scRNA-seq) technology was used to identify the cellular and molecular programs that underlie the generation of the first HSC from HE in human embryos ([66]16). However, the functional contribution of these niche cells to human hematopoiesis remains poorly defined. Single-cell transcriptional profiling has unique advantages in the identification of heterogeneous cell populations and has been successfully used to decipher the events in the development of hPSC-derived ECs, cardiomyocytes, and kidney organoid cells ([67]17–[68]19). In hematopoietic differentiation from hPSCs, HPCs and the EHT have also been analyzed at the single-cell level ([69]11, [70]20–[71]22). However, strategies for the generation of functional hematopoietic cells from hPSCs have not been identified. Here, by investigation of the entire process of pluripotent-to-hematopoietic transition, we have provided a complete transcriptome landscape of hPSC-derived hematopoiesis at the single-cell level. Through comparison of in vivo and in vitro data, we found that aerobic metabolism was increased during EHT in hPSCs, and exposure to hypoxia enhanced HPC generation with functional T cell potential from hPSCs via the induction of arterial niche signals. RESULTS scRNA-seq analysis of hematopoietic directed differentiation To gain insights into the genetic regulation of hematopoietic development, we performed single-cell transcriptional profiling of human embryonic stem cells (H1 hESCs) navigating from pluripotency through the stage-specific transitions of hematopoietic differentiation using 10× Genomics Chromium platform. A monolayer-based, chemically defined culture was modified as an efficient method to direct pluripotent cell differentiation toward the endothelial and hematopoietic (EC-HC) lineages as previously described ([72]23–[73]25). Mesoderm progenitors (T-GFP^+), CD34^+KDR^+CD144^+ ECs, and CD43^+ HPCs were identified at days 2, 4, and 6 of differentiation, respectively (fig. S1A), indicating that the period between days 4 and 6 is a key stage in EHT. For scRNA-seq analysis, we analyzed cells at time points corresponding to stage-specific transitions in the cell state including pluripotency (day 0), mesoderm specification (day 2), and progression through EC (day 4) and HPC (day 6) ([74]Fig. 1A). A total of 64,021 cells were captured of which 37,666 cells were retained after quality control analysis. Fig. 1. High-resolution dissection of hematopoietic differentiation of hPSCs using scRNA-seq. [75]Fig. 1. [76]Open in a new tab (A) Schematic diagram of hematopoietic differentiation from H1 hESCs. Cells were collected on days 0, 2, 4, and 6 for scRNA-seq. Meso, mesoderm. (B) Identification of cell populations in the differentiation culture at days 0, 2, 4, and 6 visualized by uniform manifold approximation and projection (UMAP). Each dot represents one cell, and colors represent cell clusters as indicated. (C) UMAP visualization of the expression of typical feature genes for the identification of cell clusters. (D) Expression of genes in seven different categories of the indicated cell clusters. (E) Trajectory reconstruction of all single cells throughout hematopoietic differentiation (excluding day 0) reveals three branches: prebranch (before bifurcation), Mes lineage branch, and EC-HC lineage branch. (F) Gene expression heatmap of the top 3000 highly variable genes among cells used in the trajectory inference analysis presented in a pseudo-temporal order. (G) Expression dynamics of the top 3000 highly variable genes cataloged into four major clusters in a pseudo-temporal order shown as blue lines (Mes lineage) and red lines (EC-HC lineage). Thick lines indicate the average gene expression patterns in each cluster. Gene signatures and expression dynamics of representative genes in each gene cluster are shown. (H) Gene ontology (GO) analyses of each gene cluster identified in (F) and (G). ER, endoplasmic reticulum. Unsupervised clustering of the entire pooled dataset revealed seven transcriptionally distinct populations consisting of hPSC, mesodermal progenitor cell (MPC), day 4 Mes (D4-Mes), day 6 Mes (D6-Mes), day 4 EC (D4-EC), day 6 EC (D6-EC), and HPC clusters ([77]Fig. 1B and fig. S1B). The clusters of hPSC and MPC were identified by feature gene expression (NANOG and SOX2 for hPSCs and MIXL1 and MESP1 for MPCs) ([78]Fig. 1, C and D). Compared with D4-Mes, D6-Mes were more mature with higher expression of PDGFRA and COL1A1 ([79]Fig. 1, C and D). Compared with the D6-EC cluster, the D4-EC cluster not only showed endothelial features of CDH5 (CD144), PECAM1 (CD31), and FLT1 expression but also exhibited unique hematopoietic features of RUNX1 and GATA2 expression ([80]Fig. 1, C and D). The HPC cluster was characterized by the expression of SPN (CD43), SPI1 (PU.1), and GFI1B ([81]Fig. 1, C and D). SPI1 and GFI1B are transcriptional regulators that play crucial roles in the generation of functional HPCs from hPSCs ([82]2, [83]26). Next, we established the developmental trajectories by ordering cells in a pseudo-temporal manner for the lineage reconstruction of biological processes based on transcriptional similarity. First, analysis of cells from all stages revealed that the cells progressed sequentially from the hPSC to the MPC and Mes and, lastly, to the EC and HPC lineages (fig. S1C). Then, to improve the resolution and focus on the EHT process, we constructed a differentiation trajectory following removal of the hPSC and found that MPC-derived D4-Mes (prebranch) bifurcated into two diverse branches: one branch representing EC-HC lineages and the other branch representing the mesenchymal lineage (Mes lineage) ([84]Fig. 1E and fig. S1D). Furthermore, we compared the developmental trajectories in vitro and in vivo by reanalyzing the published scRNA-seq data from human embryos ([85]16) to construct the trajectory of in vivo developmental pathway. We found that the developmental trajectory in vivo was similar to that in vitro (fig. S1E). Overall, these findings suggest that a mesenchymal-like intermediate state is involved in the development of EC-HC lineage. To gain further insights into the gene expression dynamics of the Mes and EC-HC lineages, we analyzed the expression changes of the top 3000 highly variable genes among cells collected at days 2, 4, and 6. We identified four major categories of transcriptional gene clusters in characteristic patterns ([86]Fig. 1, F to H, and fig. S1F). The genes in cluster 1, largely involved in the regulation of mesoderm (e.g., CDX1 and MESP1), were gradually down-regulated during the differentiation ([87]Fig. 1, F to H). The genes in cluster 2, enriched in cell division and cell cycle (e.g., RPS28 and CCNB1), were transiently up-regulated and then down-regulated along both mesenchymal and EC-HC lineages, representing progenitor cells with active proliferative potential ([88]Fig. 1, F to H, and fig. S1F). The genes in cluster 3, which increased their expression along mesenchymal differentiation (e.g., IGF2 and COL1A1), were transiently up-regulated and then down-regulated during EC-HC development, suggesting a biphasic regulation of mesenchymal genes during hematopoiesis as previously reported ([89]Fig. 1, F to H, and fig. S1F) ([90]27). Last, the genes in cluster 4 were activated at the later stage of differentiation with predominant involved in EC-HC development (e.g., ESM1 and SPN) ([91]Fig. 1, F to H, and fig. S1F). These data depict the trajectory of hPSC-derived hematopoietic differentiation and reveal the ordered activation of transcriptional waves throughout this process. Dissecting the heterologous cellular components involved in EHT To capture the precise cellular components involved in EHT, we extracted cell populations involved in the EC-HC lineage (including D4-EC, D6-EC, and HPC) for further analysis. By unsupervised clustering, the D4-EC population was further divided into three subclusters ([92]Fig. 2A). The smallest cluster was Mes-like with high expression of collagen genes and typical mesenchymal TFs (e.g., HAND1 and SNAI2) ([93]Fig. 2, A, B, and E) and predominant enrichment in the term of mesenchyme development ([94]Fig. 2C). The mesenchymal subpopulation in D4-EC cluster also expressed low levels of endothelial- and hematopoiesis-related genes, such as CD34 and RUNX1 ([95]Fig. 2E), with involvement in the gene ontology (GO) term “blood vessel morphogenesis” ([96]Fig. 2C). Therefore, this mesenchymal subpopulation represented a unique progenitor population with EC-HC potential, thus was named as endothelial and hematopoietic–biased Mes (EH-Mes). The largest subcluster in D4-EC cluster was endothelial progenitor cells (EPCs) given the main expression of endothelia genes (CD34 and KDR) and endothelial-regulated TFs [e.g., ID1 ([97]28)] and enrichment in blood vessel morphogenesis ([98]Fig. 2, A to C and E). The third subcluster showed high expression of HE-related TFs (e.g., RUNX1 and GATA2) ([99]Fig. 2, B and E) together with endothelial features (CD34 and KDR) ([100]Fig. 2E). This subcluster was mainly involved in the GO term “myeloid cell differentiation” ([101]Fig. 2C) but with very low expression of hematopoietic genes (SPN) ([102]Fig. 2E); therefore, this subpopulation was annotated as D4-HE. Fig. 2. Heterologous cellular components and their interactions involved in EHT. [103]Fig. 2. [104]Open in a new tab (A) UMAP visualization of EH-Mes, EPC, and D4-HE clusters based on subdivision of cells in the D4-EC cluster described in [105]Fig. 1B. (B) Heatmap showing the average expression of the top seven cluster-specific TFs among the EH-Mes, EPC, and D4-HE clusters. (C) The major GO Biological Process (GO BP) terms in which cluster-specific genes are enriched for EH-Mes, EPC, and D4-HE clusters. (D) UMAP visualization of AE, VE, D6-HE, and D6-HPC clusters based on subdivision of cells in the D6-EC and HPC clusters described in [106]Fig. 1B. (E) Violin plots showing the expression of feature genes in each cell cluster. (F) Heatmap showing the average expression of the top seven cluster-specific TFs among the AE, VE, D6-HE, and D6-HPC clusters. (G) The major GO:BP terms in which cluster-specific genes are enriched for D6-HE and D6-HPC clusters. (H) The major GO:BP terms in which cluster-specific genes are enriched for D4-HE and D6-HE clusters. ATP, adenosine triphosphate; SRP, signal recognition particle. (I) Schematic diagram showing the ligand-receptor interactions between HE or HPC and other niche clusters (AE, VE, and Mes). (J) Ligand-receptor interaction network showing the potential communications among niche cells (AE, VE, and Mes) and hematopoiesis-related cells (D6-HE and D6-HPC). (K) Heatmap showing the ligand-receptor pairs that exhibit different expression patterns among distinct niche clusters when coupled with D6-HE or D6-HPC. The previously identified HPC cluster shown in [107]Fig. 1B was further divided into two subclusters ([108]Fig. 2D). The smaller subcluster showed high expression of RUNX1 together with endothelial features, but with low expression of SPN, and was, therefore, annotated as D6-HE ([109]Fig. 2, D to G). The enrichment of GO terms in D6-HE, including translational initiation and mRNA processing ([110]Fig. 2H), was transcriptionally similar to that in the HE cluster identified in human embryos ([111]16). Compared with D4-HE, D6-HE was in a state of notable down-regulation of oxidative metabolism ([112]Fig. 2H). The other subcluster, annotated as D6-HPC, was high expression of hematopoietic genes (such as SPN and RUNX1) with low expression of endothelial genes and mainly enrichment in myeloid activation ([113]Fig. 2, D to G). The D6-EC cluster was also further divided into two subclusters ([114]Fig. 2D). Although both subclusters showed enrichment in angiogenesis and endothelial development (fig. S2A), one of the subclusters showed high expression of genes related to arterial endothelium (AE) (e.g., SOX17, GJA5, EFNB2, DLL4, HEY1, HEY2, and HES4), while the other subcluster showed high expression of genes related to venous endothelium (VE), such as NR2F2, EPHB4, NRP2, and APLNR ([115]Fig. 2, D to F, and fig. S2B). Thus, the two subclusters were identified as AE and VE, respectively. Heterologous cellular interactions of hemogenic and hematopoietic cells with niche cells To predict the cellular interactions involved in human hematopoietic development, we assessed the potential cell-cell interactions of D6-HE and D6-HPC with surrounding cellular niches (AE, VE, and D6-Mes) by ligand-receptor interaction analysis ([116]Fig. 2I). The interactions between hematopoietic-related cells and niche cells were shown to be complex and diverse ([117]Fig. 2J). When coupled with D6-HE or D6-HPC, the receptor-ligand gene pairs exhibited both similar and different expression patterns among the distinct niche populations (fig. S2C). To investigate the distinct roles of niche cells in hematopoietic development, we took the unique interactions of D6-HE and D6-HPC with niche cells into consideration ([118]Fig. 2K). Compared with VE or D6-Mes, the receptor-ligand interactions of either D6-HE or D6-HPC with AE were involved in NOTCH-related (e.g., JAG-NOTCH and DLL-NOTCH), cytokine-related (e.g., PDGFB-PDGFRB), cell adhesion–related (e.g., JAM3-JAM3), and cytotoxin-related (e.g., TNFSF10-TNFRSF10B) receptor-ligand interactions ([119]Fig. 2K and fig. S2D). Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed that the interaction between AE and D6-HE was specifically enriched in the Notch signaling pathway (fig. S2E). The interactions of either D6-HE or D6-HPC with D6-Mes were mainly enriched in integrin-related, cytokine-related, TGF-β–related, and bone morphogenetic protein (BMP)–related receptor-ligand interactions ([120]Fig. 2K). Collectively, our data establish the critical interactions of HE and HPCs with cellular niches involved in human hematopoietic development. These interactions should be further investigated to identify potential molecular mechanism of niche cells in regulating hematopoietic differentiation. Oxidative metabolism is dysregulated during in vitro directed differentiation from hPSCs We next aimed to identify differences in the activation of signals that direct hematopoiesis in vivo and in vitro, which may facilitate the development of a robust method for the generation of functional hematopoietic cells from hPSCs. By comparing the subclusters identified in our study against scRNA-seq data from human embryos ([121]16), we found the overall gene expression patterns of cells differentiated from hPSCs in vitro were similar to those in vivo ([122]Fig. 3A and fig. S3A). To establish a strategy for the efficient production of functional hematopoietic cells from hPSCs, we further compared the HE and HPC generated in vitro and in vivo by GO enrichment analysis. Our GO enrichment analysis showed that hPSC-derived HE and HPC were more active in aerobic metabolism than those during embryonic development in vivo ([123]Fig. 3, B and C, and fig. S3B). Furthermore, we compared our in vitro HPC data with the published data of hematopoietic stem and progenitor cells (HSPCs) from the human fetal liver ([124]29) and bone marrow ([125]30) and hematopoietic cells from human AGM ([126]16). In vitro HPCs showed a predominant enrichment in oxidative and mitochondrial metabolism, compared with the fetal liver, bone marrow, and AGM (fig. S3, C to E). Collectively, these data demonstrate that the early hematopoietic development in vivo occurs under hypoxia. Fig. 3. Down-regulation of oxygen metabolism at the stage of EHT. [127]Fig. 3. [128]Open in a new tab (A) Classification heatmap showing that the identified subclusters differentiated in vitro are similar to those in human embryos. The data of Carnegie stage (CS) 10/11 and CS13 cells from human embryos were reanalyzed. Hema, hematopoietic cells; Epi, epithelial cells. (B) Enriched GO terms in D6-HE and HE in vivo, respectively. (C) Enriched GO terms in D6-HPC and hematopoietic cells in vivo, respectively. (D) Volcano plot displaying the differentially expressed genes (DEGs) between cells of the EC-HC lineage on days 4 and 6. Representative respiratory chain (red) and endothelial-hematopoietic (blue) genes are indicated. Gray dots represent non-DEGs [approximately <1.6 fold change (FC)]. (E) GO analysis of down-regulated and up-regulated genes in the EC-HC lineage comparing day 6 with day 4. ncRNA, noncoding RNA; rRNA, ribosomal RNA. (F) The raw data of oxygen consumption rate (OCR) on day 4 and day 6 H1-derived CD34^+ cells determined using a Seahorse XF analyzer in the presence of the mitochondrial inhibitors (1 μM oligomycin, 5 μM carbonyl cyanide p-trifluoromethoxyphenylhydrazone, or 1.5 μM rotenone plus antimycin A). (G) Quantitation of basal, ATP-linked, and maximal OCR evaluated by two-way analysis of variance (ANOVA) (n = 5). (H) The raw data of glycolytic proton efflux on day 4 and day 6 H1-derived CD34^+ cells following injection of rotenone plus antimycin A (0.5 μM) and 2-deoxy-d-glucose (50 mM); data are obtained using a Seahorse XF analyzer. (I) Quantitation of basal and compensatory glycolysis evaluated by two-way ANOVA (n = 5). Data are presented as means ± SEM. **P < 0.01 and ***P < 0.001. Considering that D6-HE are in a relatively hypoxic metabolic state compared with D4-HE ([129]Fig. 2H), we speculated that the oxygen level may be lowered during EHT. To further investigate the molecular changes during EHT, we analyzed the differentially expressed genes (DEGs) between cells in the EC-HC lineage on days 4 and 6. In comparison with the day 4 EC-HC lineage, the up-regulated genes in the day 6 EC-HC lineage were strongly enriched for EC-HC development, while the down-regulated genes involved in the day 6 EC-HC lineage were enriched for GO terms related to oxygen consumption, including typical respiratory chain genes, such as UQCRQ, NDUFC1, COX17, MRPL52, and TIMM8B ([130]Fig. 3, D and E). To confirm the metabolic status during EHT, we used a Seahorse XF analyzer to measure the oxygen consumption rate (OCR) of CD34^+ EC-HC cells on days 4 and 6. Compared with day 6 CD34^+ cells, a significant increase in mitochondrial respiration was observed in day 4 CD34^+ cells ([131]Fig. 3, F and G). Using glycolytic rate assay to determine the glycolytic proton efflux rate of CD34^+ cells on days 4 and 6, we observed a significant increase in glycolysis in day 6 CD34^+ cells ([132]Fig. 3, H and I). Collectively, these results suggest that, although hematopoietic differentiation in vitro occurs in a relatively aerobic metabolic state, down-regulation of oxygen metabolism is still required to drive EHT and generate HPCs from HE cells generated in vitro. Hypoxia improves hematopoietic differentiation of hPSCs by activation of the arterial specification program To investigate the role of hypoxia in hematopoietic differentiation from hPSCs, we cultured cells under conditions of hypoxia with 5% O[2] for 6 days of differentiation. Treatment of the differentiated H1 hESCs with hypoxia enhanced the formation of semiadhesive round cell clusters and increased the number of CD43^+ HPCs on day 6 ([133]Fig. 4, A and B, and fig. S4, A and B). The enhanced effect of hypoxia on hematopoietic differentiation was also confirmed in human induced pluripotent stem cells (iPSCs) (JHUi181 and BC1), which showed similar results to the H1 hESCs ([134]Fig. 4B). Quantitative real-time polymerase chain reaction (qRT-PCR) analyses of the differentiated H1 hESCs showed that the expression of key genes associated with early hematopoiesis—including MYB, RUNX1, TAL1, and GATA2—was significantly increased under hypoxic conditions than that under normoxic conditions (fig. S4C). The colony-forming cell (CFC) assay of the differentiated H1 hESCs showed that hypoxia-induced hematopoietic differentiation gave rise to significantly higher numbers of hematopoietic colonies of myeloid and erythroid lineages than those generated under normoxia (fig. S4D), indicating that hypoxia enhanced the development of multipotent HPCs. To examine the hypoxia-dependent stage more precisely, we applied hypoxic conditions for defined intervals during the 6 days of hematopoietic differentiation of H1 hESCs ([135]Fig. 4C). As expected, applying hypoxia with 1% O[2] consistently for 6 days significantly increased the frequency of CD43^+ hematopoietic cells ([136]Fig. 4D). When hypoxia was applied between days 4 and 6, the stage coinciding with onset of EHT, the frequency of CD43^+ hematopoietic cells was significantly increased, compared with other 2-day interval groups ([137]Fig. 4D). Collectively, these findings demonstrate that hypoxia enhances hematopoietic differentiation of hPSCs at the stage of EHT. Fig. 4. Hypoxia enhances hematopoietic differentiation by promoting the arterial specification program. [138]Fig. 4. [139]Open in a new tab (A) Confocal microscopy visualization of emerging round hematopoietic cells on day 6 under the hypoxic (5% O[2]) or normoxic conditions. Scale bars, 40 μm. (B) The percentage of CD43^+ hematopoietic cells on day 6 under the hypoxic (5% O[2]) or normoxic conditions. (n = 3; two-way ANOVA). (C and D) Diagram and representative flow cytometric analysis demonstrating the five conditions used to test the importance of hypoxia exposure (1% O[2]) in promoting hematopoiesis. (n = 3). (E) Mean gene expression of all cells at each time point. (F) HIF-1a expression measured by Western blot at each time point under the normoxic or hypoxic conditions (1% O[2]). (G and H) Representative flow cytometric and statistical analyses of the dynamic changes in the arterial cell population between days 4 and 6 under hypoxic (1% O[2]) or normoxic conditions. (n = 3; two-way ANOVA). (I) qRT-PCR analysis of the expression of DLL4 and NR2F2 from days 4 to 6 under the hypoxic (1% O[2]) or normoxic conditions. (n = 3; two-way ANOVA). (J) Representative flow cytometric analysis of the frequency of arterial cells in dimethyl sulfoxide (DMSO)– or U0126-treated day 5 cells. (n = 3). (K and L) Flow cytometric analysis of the frequency and number of CD43^+ cells in DMSO- or U0126-treated day 6 cells. (n = 3; two-tailed Student’s t test). SSC, side scatter. For (J) to (L), U0126 or DMSO was added to the cultures from days 4 to 6 of differentiation. Experiments were performed on H1 unless otherwise indicated. Data are presented as means ± SEM. *P < 0.05 and ***P < 0.001. The vascular niche plays a key role in the specification and amplification of HSCs ([140]13, [141]26, [142]31). Further analysis of gene expression during EHT revealed that the expression of hypoxia gene HIF-1a and its related activated genes increased on day 4, peaked on day 5, and then decreased ([143]Fig. 4E and fig. S4E). The high HIF-1a expression in mRNA on day 0 may be due to the anaerobic metabolism of hPSCs ([144]32). The increased expression of HIF-1a and its related activated genes under normoxic and hypoxic conditions during EHT was further confirmed by Western blot and qRT-PCR, respectively ([145]Fig. 4F and fig. S4F). Compared with HIF-1a expression, the endothelial-related genes showed similar expression patterns ([146]Fig. 4E). Compared with venous genes (NR2F2 and EPHB4), arterial genes (SOX17 and EFNB2) were notably up-regulated on day 4 and peaked on day 5, accompanied by a notable increase in NOTCH-related genes ([147]Fig. 4E and fig. S4G). Therefore, we speculated that the effect of hypoxia on hematopoiesis is related to the activation of arterial program. To dynamically track the effect of hypoxia on the activation of arterial program during EHT, we assessed cell populations with the expression of CD73 and CD184, which have been used to distinguish arterial cells (CD34^+CD43^−CD73^+CD184^+ AE and CD34^+CD43^−CD73^−CD184^+ HE with arterial features) from CD34^+CD43^−CD73^+CD184^− VE ([148]33–[149]35). Compared with days 4 and 6, it was interesting to see that VE were decreased under hypoxic condition on day 5, whereas arterial cells, including AE and HE, were increased significantly on day 5 ([150]Fig. 4, G and H, and fig. S4, H to J). Using qRT-PCR, we found that hypoxia effectively up-regulated the gene expression of DLL4, a key marker of arterial cells, and down-regulated the gene expression of NR2F2, a key marker of VE ([151]Fig. 4I). Compared to the CD34^+CD43^−CD73^−CD184^+ HE cell population, the CD34^+CD43^−CD73^−CD184^− cell population has also been shown to have the ability to undergo EHT to generate HPCs but with limited T cell differentiation potential ([152]9, [153]33, [154]35). The modest decrease in the CD34^+CD43^−CD73^−CD184^− cell population and significant increase in the CD34^+CD43^−CD73^−CD184^+ HE cell population on day 5 under hypoxic conditions ([155]Fig. 4G and fig. S4K) suggested that hypoxia enhanced multipotent HPC generation. To further determine whether the effect of hypoxia on hematopoiesis is arterial program dependent, we applied a mitogen-activated protein kinase (MAPK) kinase 1 (MEK1) and MEK2 inhibitor, U0126, to inhibit MAPK/extracellular signal–regulated kinase (ERK) signaling pathway that was involved in the development of arterial program ([156]33, [157]35, [158]36). We found that inhibition of MEK/ERK between days 4 and 6 impaired the development of arterial cells under hypoxia with 1% O[2] and led to a significant decrease in the proportion of CD43^+ HPCs ([159]Fig. 4, J to L, and fig. S4L). Overall, these observations demonstrate that hypoxia enhances hematopoietic differentiation of hPSCs by promoting the arterial specification program. AE promote the generation of hematopoietic progenitors with T cell potential Since arterial program includes AE and arterial-type HE ([160]9, [161]35), it is still unclear whether hypoxia effect on hematopoietic differentiation is through cell-autonomous niches, cellular niches, or both. To further determine the roles of arterial cells and other cellular niches in hematopoiesis, AE (CD34^+CD43^−CD73^+CD184^+), VE (CD34^+CD43^−CD73^+CD184^−), HE (CD34^+CD43^−CD73^−CD184^+), or Mes (CD34^−CD31^−CD90^+CD105^+) were isolated from day 5 differentiated H1 hESCs and induced EHT for 7 days (D5 + D7) with or without coculture ([162]Fig. 5A and fig. S5A). During EHT, AE, VE, or Mes alone was unable to generate CD43^+ and CD45^+ hematopoietic cells, indicating that the absence of hematopoietic potential in these populations (fig. S5B). Compared with the coculture of HE cells with VE (HE + VE) or Mes (HE + Mes), HE cells alone were unable to initiate EHT efficiently, whereas the coculture of HE cells with AE (HE + AE) for 7 days promoted EHT, significantly increasing the frequency and total numbers of CD43^+ and CD45^+ HPCs ([163]Fig. 5, B and C). The AE niche effect on HPC generation from HE cells was also confirmed in BC1 human iPSCs, which showed similar results to the H1 hESCs (fig. S5C). Overall, these data demonstrate that hypoxia enhances hematopoiesis in two ways: (i) increasing arterial-type HE and (ii) promoting EHT by AE cellular niche. Fig. 5. AE promote hematopoietic progenitor generation with T cell potential. [164]Fig. 5. [165]Open in a new tab (A) Schematic of protocol for investigating the niche effect on hematopoiesis. (B) Representative flow cytometric analysis of the hematopoietic progenitors obtained from H1-derived HE, or HE cocultured with Mes (HE + Mes), VE (HE + VE), or AE (HE + AE) following 7 days of EHT culture (n = 3). (C) The fold change in CD43^+CD45^+ cell number compared to HE. The cells were generated under the conditions of HE, HE + Mes, HE + VE, or HE + AE. (n = 3; one-way ANOVA). (D) CFC assay of HPCs generated with or without coculture with AE, VE, or Mes. Colony-forming units (CFUs) per 2000 cells plated. (n = 3; two-way ANOVA). (E) Schematic diagram of the protocol for T cell differentiation. MACS, magnetic-activated cell sorting. (F and G) T cell potential of hematopoietic progenitors generated from D5 HE following 7 days of culture in the presence or absence of VE, AE, or Mes. (n = 3; one-way ANOVA). (H) Representative flow cytometric analysis of the expression of CD4 and CD8 by CD3^+ cells before and after anti-CD3/CD28 stimulation (n = 3). (I) The proportion of CD107a in hPSC-derived T cells (hPSC-T) and peripheral blood T cells (PB-T) after treatment with phorbol 12-myristate 13-acetate (PMA)–ionomycin. (n = 3; two-way ANOVA). (J) Polyfunctional cytokine production by hPSC-T and PB-T after treatment with PMA-ionomycin. (n = 3; two-way ANOVA). Experiments were performed on H1 hESCs unless otherwise indicated. Data are presented as means ± SEM. *P < 0.05 and ***P < 0.001. To determine the multilineage potential of HPCs, we harvested hematopoietic cells in suspension after EHT induction with or without coculture with niche cells (HE, HE + VE, HE + Mes, or HE + AE) and measured their colony forming potential. The total number of colonies was significantly increased in the HE + AE cultures, while there were no significant differences in the total number of colonies between the HE cultures and the HE + VE or HE + Mes cultures ([166]Fig. 5D). These results suggest that AE niche not only promotes EHT but also increases multilineage potential of emerged HPCs. To determine whether the effect of AE niche on hematopoiesis is Notch dependent, we applied the Notch inhibitor, DAPT, to inhibit the Notch signaling in AE. We found that inhibition of Notch in HE + AE cocultures led to a significant decrease in the proportion of CD45^+ cells (fig. S5D). T cell potential has been identified as a hallmark of multipotent HPCs ([167]9, [168]33, [169]35, [170]37). To further investigate the effect of cellular niches on the T cell potential of HPCs, we induced T cell differentiation from H1 hESCs using a previously reported artificial thymic organoid (ATO) model involving coculture of CD45^+ HPCs with OP9–human delta-like 1 (hDLL1) cells in three-dimensional (3D) aggregates for 8 weeks ([171]Fig. 5E) ([172]38, [173]39). We found that the isolated CD45^+ HPCs derived from AE cocultures (HE + AE) gave rise to increased CD3^+ T cell numbers, whereas the T cell potential of CD45^+ HPCs from VE or Mes cocultures, or without niche cells, was limited ([174]Fig. 5, F and G). The CD3^+ T cells exhibited a naïve phenotype with the expression of αβ T cell receptor (TCR-αβ), CD45RA, CD62L, CD28, and CD27 (fig. S5E). Before anti-CD3/CD28 treatment, CD8^+CD4^− cells and CD8^+CD4^+ cells were included among the CD3^+ cells ([175]Fig. 5H). After anti-CD3/CD28 treatment, CD8^+CD4^− cells accounted for more than 90% of the CD3^+ cells ([176]Fig. 5H). To examine whether the T cells derived from AE-primed HPCs in vitro are functional, we stimulated the CD3^+ cells in the presence of the T cell activator phorbol 12-myristate 13-acetate (PMA)–ionomycin ([177]Fig. 5E) and quantitated surface marker expression and cytokine production. After stimulation, CD25^+CD69^+ T cells significantly increased (fig. S5, F and G). Similar to peripheral blood (PB) T cells (PB-T), the hPSC-derived T cells (hPSC-T) generated from AE-primed HPCs were highly functional, releasing cytotoxic granules (CD107a expression) ([178]Fig. 5I and fig. S5H) with polyfunctional production of interferon-γ (IFN-γ), tumor necrosis factor–α (TNF-α), and interleukin-2 (IL-2) ([179]Fig. 5J). Overall, these data demonstrate that AE promote the generation of HPC with functional T cell potential from HE cells. AE-primed hPSC-T showed therapeutic potential following engineered expression of anti–CD19 chimeric antigen receptor To further evaluate the function and the potential application of T cells derived from AE-primed HPCs, the T cells were engineered to express anti-CD19 chimeric antigen receptor (hPSC-CAR-T) for cytotoxicity assays both in vitro and in vivo ([180]Fig. 6A). The conventional CAR-T constructed from PB-isolated T cells (PB-CAR-T) were used as a positive control; hPSC-T and PB-T transfected with empty vectors (hPSC-VEC-T and PB-VEC-T) were used as negative controls. The CD19 CAR infection efficiency for hPSC-T was between 60 and 80%, which was similar to that of PB-T (fig. S6A). CD19^+ cell lines (Nalm-6 and Raji) and a CD19^− cell line (Molm13) were used as target cells (fig. S6B). hPSC-CAR-T and PB-CAR-T showed significant cytotoxic granule release (CD107a expression) when cocultured with Nalm-6 and Raji cells, while little CD107a up-regulation was observed following coculture with Molm13 cells ([181]Fig. 6B). After 24 hours of coculture, proinflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA). The results showed that the secretion of IL-2, TNF-α, and IFN-γ was significantly increased in both hPSC-CAR-T and PB-CAR-T when cocultured with CD19^+ target cells ([182]Fig. 6C). By performing true lytic capability assays, we further confirmed in vitro that the hPSC-CAR-T killed CD19^+ leukemia cells as effectively as PB-CAR-T ([183]Fig. 6D). Fig. 6. AE-primed hPSC-T showed therapeutic potential toward B-ALL following engineered expression of anti–CD19-CAR. [184]Fig. 6. [185]Open in a new tab (A) Schematic diagram showing the protocol for generating hPSC-CAR-T for cytotoxicity assays both in vitro and in vivo. The AE used to prime T cells are generated under the hypoxic condition (1% O[2]). (B) The proportion of CD107a^+ cells in effector cells after coincubation with target cells (E:T = 1:1) for 5 hours. (n = 3; two-way ANOVA). (C) ELISA data showing the release of IFN-γ, IL-2, and TNF-α after coincubation with target cells for 24 hours. (n = 3; two-way ANOVA). (D) Direct lysis of effector cells against target cells. Flow cytometry analysis of the percentage of CD19^+CD3^− cells. (n = 3; two-way ANOVA). (E and F) Representative bioluminescence imaging and statistical analysis of the bioluminescence intensity in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. (n = 7; two-way ANOVA). (G) Body weight of the mice. (n = 7; two-way ANOVA). (H) Kaplan-Meier curve representing the percentage survival of the mice. (n = 7; log-rank test). (I) Specific fluorescence intensity (SFI) of CD19 on the bone marrow mononuclear cells (BMMNCs) in six patients with B acute lymphoblastic leukemia (B-ALL). (J) The proportion of CD107a^+ cells in effector cells after coincubation with primary B-ALL target cells at E:T = 1:1 for 5 hours. (n = 6; one-way ANOVA). (K) The release of IFN-γ, IL-2, and TNF-α after coincubation with primary B-ALL target cells for 24 hours. (n = 6; one-way ANOVA). (L) Direct lysis of effector cells against primary B-ALL target cells. (n = 6; two-way ANOVA). All values are means ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001. To further investigate the cytotoxic efficacy of hPSC-CAR-T toward CD19^+ B acute lymphoblastic leukemia (B-ALL) in vivo, we established a B-ALL mouse model as previously described ([186]40). A total of 3 × 10^5 luciferase-expressing Nalm-6 cells (Nalm-6–luc2) were intravenously injected into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. At days 4 and 11, 5 × 10^6 PB-VEC-T, hPSC-VEC-T, PB-CAR-T, or hPSC-CAR-T were administered intravenously ([187]Fig. 6A). Bioluminescent imaging showed that the injection of hPSC-CAR-T inhibited tumor progression ([188]Fig. 6, E and F), delayed weight loss ([189]Fig. 6G), and prolonged the survival of the treated mice ([190]Fig. 6H). Using the same protocol, we showed that the therapeutic performance of hPSC-CAR-T and PB-CAR-T was comparable ([191]Fig. 6, E to H). Last, we compared the cytotoxicity of hPSC-CAR-T and PB-CAR-T in primary B-ALL cells. Bone marrow mononuclear cells (BMMNCs) collected from six patients with B-ALL (table S1) were used as target cells. The CD19 expression level was detected ([192]Fig. 6I and fig. S6C). We showed that hPSC-CAR-T and PB-CAR-T shared similar cytotoxic characteristics against CD19^+ target cells, including degranulation ability ([193]Fig. 6J), cytokine release ([194]Fig. 6K), and true lytic capability ([195]Fig. 6L). These data demonstrate that, through engineered expression of anti–CD19-CAR, the hPSC-T generated from AE-primed HPCs potently inhibit the progression of leukemia both in vitro and in vivo. DISCUSSION In the present study of hPSC differentiation, we constructed the first genome-scale gene expression landscape covering the entire course of the pluripotent-to-hematopoietic transition at the single-cell level. To avoid artificial bias and more precisely track hematopoietic fate choices, no cell sorting or marker enrichment was performed before capture of the single cells. By constructing the cellular and molecular expression profiles of human early hematopoiesis, we identified decreased oxygen metabolism as a previously unidentified molecular driver of EHT. Functionally, we showed that hypoxia enhanced hematopoietic differentiation of hPSCs by activating the arterial specification program that governed the generation of HPCs with committed T cell potential. The T cells generated from AE-primed HPCs showed a strong tumor killing potential both in vitro and in vivo following engineered expression of anti–CD19-CAR. A recent study of EHT has shown that HE cells are maintained in a quiescent state and reentry into the cell cycle is required to initiate EHT ([196]11). Our study confirmed active cell division before EHT ([197]Fig. 1, G and H, and fig. S1F). Furthermore, we revealed that once EHT occurred, gene expression by the cells was gradually silenced ([198]Fig. 1G), suggesting that a biphasic regulation of the cell cycle plays an important role in EHT. A previous report of the biphasic regulation of mesenchymal genes during EHT ([199]27) was also confirmed in our study at the single-cell level. We demonstrated that the expression of mesenchymal genes was up-regulated before EHT and attenuated during EHT ([200]Fig. 1, G and H, and fig. S1F). Beyond these initial findings, we demonstrated that oxidative metabolism played an important role in EHT ([201]Fig. 3, B to I). The effects of hypoxia on the maintenance and differentiation of adult HSCs have been widely studied ([202]41). However, because of the limitation of resources and ethical issues, little is known about the role of hypoxia in regulating early human hematopoiesis. In our study, we not only found that the process of EHT was accompanied by a decrease in oxygen metabolism but also showed that hypoxia enhanced HPC generation by promoting the arterial specification program. The mechanism by which hypoxia promotes arterial specification is not well characterized. However, a study of mouse embryonic stem cell differentiation indicated that hypoxia first induced the generation of EC progenitors by up-regulating the TF Etv2 and subsequently driving maturation to an AE fate via a Notch1-dependent signaling mechanism ([203]42). It is well-known that HE cells are derived from a subset of ECs; however, where and how the ECs with hematopoietic potential originate have not yet been fully elucidated. By dissecting the heterologous cellular components and their trajectories involved in EHT in this study, we provided evidence that the EC-HC lineages originated from early progenitors with mesenchymal features ([204]Fig. 1E). Furthermore, a small group of mesenchymal progenitors with EC-HC potential, designated EH-Mes, was identified in the D4-EC cluster ([205]Fig. 2, A to C and E). A similar population, designated mesenchymoangioblast (MB) cells, with endothelial and mesenchymal potential has also been identified in a study of hPSC differentiation ([206]43). In contrast to the ECs derived from EH-Mes, MB-derived ECs are capable of generating mesenchymal stem cells but not hematopoietic cells ([207]44). Because HSC–potential HE is largely associated with arteries ([208]45–[209]47), it has been assumed that this is represented by a subpopulation of arterial vascular ECs. This hypothesis was supported by a recent transcriptomic study of human embryonic hematopoietic development demonstrating that RUNX1 was exclusively expressed in a small population of cells in the AE cluster and characterization of RUNX1^+ HE cells revealed clear arterial features ([210]16). In our study, the D6-HE was transcriptionally similar to the HE cluster identified in human embryos and was more similar to arterial HE than the D4-HE. Arterial-type HE cells were also identified in a hPSC differentiation model with the CD34^+CD144^+CD43^−CD73^−CD184^+ phenotype, and activation of the arterial specification program in HE cells was shown to drive the development of hematopoiesis with more T cell potential ([211]9, [212]35). Therefore, the arterial-type HE cells with CD34^+CD43^−CD73^−CD184^+ phenotype were applied in our functional studies ([213]Fig. 5A). It has been reported that the EHT of arterial-type HE cells is Notch dependent ([214]9, [215]35), which may explain why it is difficult to promote EHT in arterial-type HE cells without any cellular niche support ([216]Fig. 5B). We also conducted cell-cell interaction analysis to investigate the potential mechanisms by which the AE niche regulates hematopoiesis ([217]Fig. 2K and fig. S2, C to E). Notch signaling was validated as a critical regulator (fig. S5D); however, considering the many other interactions (e.g., PDGFB-PDGFRB) between AE and HE, it can be speculated that while Notch activation is required, it may not be sufficient for the maintenance of AE function. For example, we showed that PDGFB improved human HSC engraftment in our previous study ([218]48). Although the Mes and VE identified in our study have neither hematopoietic potential nor the ability to promote EHT, they may play a key role in the development of HE cells. The interaction between EC and Mes has been demonstrated to control the generation of HE cells with RUNX1 expression ([219]49). Instead of generation of CAR-T from CAR-expressing iPSCs as previous reports ([220]50, [221]51), the CAR-T in our study were done by producing T cells from hPSCs that were then transduced with the CAR construct, due to a concern that CAR expression in hPSCs may alter EHT and impair T cell differentiation. The iPSCs derived from T cells (T-iPSCs) have been reported with a higher efficiency in T cell differentiation than the non–T cell–derived iPSCs (non–T-iPSCs) ([222]52). It will be interesting to investigate in the future whether T-iPSCs and non–T-iPSCs (or hESCs) have same or different efficiencies in HPC generation and its differentiation to different hematopoietic lineages. Moreover, the function of the hematopoietic cells generated from T-iPSCs and non–T-iPSCs (or hESCs) may be different. In our study, the H1-derived T cells highly expressing CD45RA, CD62L, CD28, and CD27 (fig. S5E) are closest to naïve T cells and T memory stem cells ([223]53, [224]54) and show a strong ability to eliminate leukemia cells following engineered expression of anti–CD19-CAR.It has been reported that naïve T cells and T memory stem cells have stronger proliferation and differentiation ability when stimulated by antigen and produce greater antitumor activity than the PB-T ([225]53, [226]54). This may explain our observation of the better effects of hPSC-CAR-T than PB-CAR-T in vivo. The better effects of hPSC-CAR-T are also consistent with a previous study that TCR-transduced iPSC-derived T cells (iPSC-TCR-T) showed better effects in inhibiting tumor progression and extending the survival of mice than the PB-isolated TCR-T ([227]55). As a growing number of studies have revealed multiple overlapping waves of hematopoietic progenitors generated from HE cells ([228]56, [229]57), it is unclear whether the AE-primed CD45^+ HPCs with T cell potential are a transient wave of HSCs or T-biased definitive progenitors. In summary, we construct comprehensive cellular and molecular networks toward hematopoiesis of hPSCs at the single-cell level, which provide benchmark datasets to understand the origins of human hematopoiesis. Moreover, finding the important role of the AE niche function in lymphoid development from hPSCs provides an approach that significantly improves T cell progenitor production and facilitates advances in the translation of hPSC-based immunotherapies to the clinic. MATERIALS AND METHODS Study design The goal of this study was to investigate the key cellular and molecular signals involved in human early hematopoiesis and identify strategies for functional hematopoietic cell generation from hPSCs. We performed extensive single-cell transcriptomic analyses to map fate choices and gene expression patterns during hematopoietic differentiation of hPSCs. To identify differences in the activation of signals that direct hematopoiesis in vivo and in vitro, we compared the transcriptomic differences during early hematopoiesis in hPSCs and human embryos. For functional study, a range of in vitro assays using cell lines and primary B-ALL cells and in vivo assays using a mouse model of B-ALL was performed. For in vitro assays, at least three independent experiments were performed. We obtained samples of patients with B-ALL from the Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences. Informed consent was obtained from all participants in accordance with the Declaration of Helsinki. The procedure was approved by the ethical advisory board of the Institute of Hematology and Blood Diseases Hospital. Patient-related information is presented in table S1. For in vivo studies, female and age-matched animals were randomly assigned to experimental or control groups. The number of animals used in each study group is specified in the figure legends. All animal studies were performed according to procedures and protocols approved by the Institutional Animal Care and Use Committee of Peking Union Medical College. Maintenance and differentiation of hPSCs The hESC (H1) and hiPSC (BC1, BC1-T, and JHUi181) lines were maintained on Matrigel-coated plates with E8 medium (Life Technologies) as described previously ([230]23–[231]25). For differentiation, single hPSCs were obtained for sequential EC-HC induction, as described previously ([232]23–[233]25) with minor modifications. Briefly, single hPSCs were plated at an optimized density at 6 × 10^3 cells per well onto vitronectin (PeproTech)–coated 12-well plates in the STEMdiff APEL 2 Medium (STEMCELL Technologies) supplemented with 3 μM glycogen synthase kinase 3 inhibitor CHIR99021 [Applied Biological Materials (ABM)], activin A (2 ng/ml; PeproTech), BMP4 (10 ng/ml; PeproTech), or 10 μM rho kinase inhibitor Y-27632 (STEMCELL Technologies) on day 0. After 48 hours (day 2), the medium was changed to STEMdiff APEL Medium supplemented with vascular endothelial growth factor (VEGF; 40 ng/ml; PeproTech). On day 3, fibroblast growth factor 2 (FGF2; 40 ng/ml; ABM) was added to the cultures without aspirating the old medium. From day 4, the medium was changed to STEMdiff APEL 2 Medium supplemented with VEGF (40 ng/ml; PeproTech) and FGF2 (40 ng/ml; ABM) until day 6. Cultures were maintained at 37°C under normoxic (room air with 5% CO[2]) or hypoxic (1% or 5% O[2]) conditions as indicated. Where indicated, a MEK inhibitor (U0126; 3 μM; Selleck Chemicals) was included. T cell differentiation For OP9-hDLL1 generation, OP9 cells were transduced with a lentiviral vector encoding full-length hDLL1. The highest 5% DLL1-expressing cells were sorted by fluorescence-activated cell sorting (FACS) using an anti-DLL1 antibody and passaged in minimum essential medium–α/10% fetal bovine serum (FBS). Stable expression was confirmed by flow cytometric and qRT-PCR analyses of DLL1 expression after several weeks of culture. For T cell differentiation, 1 × 10^6 OP9-hDLL1 cells were centrifuged with 5 × 10^5 CD45^+ cells to form small 3D aggregates, which were then plated onto a 0.4-mm Millicell transwell insert (EMD Millipore) placed in a six-well plate containing 1 ml of T cell differentiation medium consisting of RPMI 1640 (Gibco), 4% B27 supplement (Thermo Fisher Scientific), 30 mM l-ascorbic acid 2-phosphate sesquimagnesium salt hydrate (Sigma-Aldrich), 1% penicillin/streptomycin (Thermo Fisher Scientific), stem cell factor (20 ng/ml; PeproTech), FLT3L (5 ng/ml; PeproTech), and IL-7 (5 ng/ml; PeproTech). Medium was changed every 3 to 4 days, and the cells were cultured for 8 weeks before harvesting for CD3^+ enrichment, followed by a further 1 week of stimulation with anti-CD3/CD28 beads (Thermo Fisher Scientific). Cells were harvested by adding TrypLE (Gibco) and disaggregating the 3D aggregate by pipetting, followed by passage through a 50-mm nylon strainer. The cells were then analyzed by flow cytometry on the days indicated. In vitro function studies of hPSC-CAR-T Nalm-6, Raji, and Molm13 cells were maintained in RPMI-1640 medium supplemented with 10% FBS (Gibco). T cells were cultured in X-VIVO 15 medium (Lonza) supplemented with 5% FBS and recombinant human IL-2 (50 to 100 U/ml; PeproTech) (T cell culture medium). Primary BMMNCs were isolated from patients and cultured as previously described ([234]58). Analysis of direct cytotoxicity hPSC-CAR-T, PB-CAR-T, hPSC-VEC-T, or PB-VEC-T were incubated with target cells at an effector-to-target (E:T) ratio of 1:1 in T cell culture medium without rhIL-2. After 8 and 24 hours, the cells were harvested and stained with phycoerythrin (PE)–conjugated anti-human CD19 antibody (BioLegend, catalog no. 302208, HIB19) and allophycocyanin (APC)-Cy7–conjugated anti-human CD3 antibody (BioLegend, catalog no. 300318, HIT3a) for 30 min at 4°C. The cells were then washed and resuspended in phosphate-buffered saline for flow cytometry analysis. The percentage of CD3^−CD19^+ cells represented the residual level of target cells. Cytokine-releasing assay hPSC-CAR-T, PB-CAR-T, hPSC-VEC-T, or PB-VEC-T (2 × 10^5) were cocultured with target cells at E:T ratios of 1:1 in 1 ml of T cell culture medium without rhIL-2 for 24 hours. After 24 hours, IL-2, IFN-γ, and TNF-α in the culture supernatants were detected using ELISA kits (R&D Systems, USA) according to the manufacturer’s instructions. Degranulation assay hPSC-CAR-T, PB-CAR-T, hPSC-VEC-T, or PB-VEC-T (1 × 10^5) were cocultured with 1 × 10^5 target cells in 200 μl of T cell culture medium with PE-conjugated anti-CD107a antibody and 100 U of rhIL-2. After 1 hour, monensin (100 μg/ml) was added into the cocultured system, and the cells were incubated for 4 hours. The cells were then labeled with APC-Cy7–conjugated anti-human CD3 antibody and analyzed by flow cytometry. All CD107a^+ cells in GFP^+ cells were regarded as degranulated T cells. In vivo NOD/SCID murine studies NOD/SCID female mice (aged 8 weeks) were purchased from the Institute of Laboratory Animal Sciences (Chinese Academy of Medical Sciences and Peking Union Medical College, China). All animal experiments were approved by the Institutional Animal Care and Use Committee of Peking Union Medical College. Mice (n = 24) were intravenously inoculated with 3 × 10^5 Nalm-6–luc2 cells. Four days after transplantation, mice were randomly divided into four treatment groups according to the average radiance of bioluminescent imaging: hPSC-CAR-T group, PB-CAR-T group, hPSC-VEC-T group, and PB-VEC-T group. The mice in these groups received intravenous injection of 5 × 10^6 hPSC-CAR-T, PB-CAR-T, hPSC-VEC-T, and PB-VEC-T, respectively, on days 5 and 10. Bioluminescent images were obtained using Caliper IVIS Lumina II (Caliper Life Sciences, USA), and the average radiance was calculated as described previously ([235]59). Statistical analysis Analyses were performed using GraphPad Prism 6.0 software. Survival rates were analyzed by the Kaplan-Meier method. Data obtained from multiple experiments were reported as the means ± SEM. An unpaired t test was used to compare the means from two groups, and analysis of variance (ANOVA) was used to compare the means from three or more groups. Results with a value of P < 0.05 were considered statistically significant. *P < 0.05, **P < 0.01, and ***P < 0.001. Acknowledgments