Mouse models
Tet2 KO (strain name: B6(Cg)-Tet2tm1.2Rao/J) parental mice were ordered from the Jackson Laboratory and crossed with Tet2WT C57BL/6J mice to heterozygous (Tet2HET) mice. The Tet2HET mice were then crossed to generate Tet2WT and Tet2KO animals in the same litter. CD45.1+ Tet2WT mice in the C57BL/6J background (strain name: B6.SJL-Ptprca Pepcb/BoyJ) were ordered from the Jackson Laboratory.
Generation of the HPCHOXB4 cell models
Bone marrow cells were isolated from 8-week-old male Tet2WT and Tet2KO littermates from a Tet2HET x Tet2HET breeding pair. Sca-1+ HSPCs were enriched using the EasySep™ mouse SCA1 positive selection kit (STEMCELL, #18756). Enriched cells were then infected with retroviral particles expressing HOXB4-Venus (a gift from Norman Iscove’s lab). Briefly, Tet2WT or Tet2KO cells were seeded at a density of 2x105 cells/mL in a well in a 24-well TC-treated tissue culture plate (Falcon, #353047) and infected with 50uL of viral supernatant diluted in 450uL of growth medium (see ‘Tissue culture’ section) in the presence of 8ug/mL of polybrene (Millipore, #TR10039). Growth medium was replaced on the next day. The level of Venus fluorescence was measured by flow cytometry at 72 hours post infection to access transduction efficiency.
Human mobilized peripheral blood (mPB) samples
Human mPB samples were obtained with informed consent from Princess Margaret Cancer Centre following the procedures approved by the University Health Network (UHN) research ethic board. Mononuclear cells were isolated by density gradient centrifugation using Ficoll-Paque™ (Fisher Scientific, #45001749), and hematopoietic progenitors were enriched through lineage marker depletion (STEMCELL, #14056).
Human cord blood samples
Human cord blood samples were obtained with informed consent from Princess Margaret Cancer Centre following the procedures approved by the University Health Network (UHN) research ethic board. Mononuclear cells from 1–5 pooled female and male donors were isolated and enriched for HSPCs using the above-mentioned protocols.
Tet2 genotyping
Genomic DNA was extracted from mouse tails and the HPCHOXB4 cell lines using standard protocols. Genotyping PCR was performed according to the protocol provided by the Jackson Laboratory for B6(Cg)-Tet2tm1.2Rao/J mice. The sequence of the primers used are shown in Table S1. Genotyping PCR was conducted using Taq polymerase (NEB, #M0495S) and buffer (ThermoFisher, #18067017). PCR products were visualized on 1.5% (w/v) agarose gels with SYBR™ Safe DNA gel stain (Thermo Scientific, #S33102).
Tissue culture
HPCHOXB4 cells were cultured in IMDM medium (Wisent, #319-105-CL) containing 10% FBS (Wisent, #080–150), murine stem cell factor (mSCF, 100ng/mL; STEMCELL, #78064.2), recombinant human FLT3 ligand (FLT3-L, 50ng/mL; STEMCELL, #78009.2), and recombinant human thrombopoietin (TPO, 25ng/mL; STEMCELL, #78210.2).
Hematopoietic progenitor cells were isolated from human mPB samples using the EasySep™ human progenitor cell enrichment kit (STEMCELL, #19356) and cultured in StemSpan SFEM II medium (STEMCELL, #09655) supplemented with 20% BIT 9500 (STEMCELL, #09500), 1% Gluta-Plus (Wisent, #609-066-EL), mSCF (100ng/mL), FLT3-L (100ng/mL), TPO (100ng/mL), StemReginin/SR1 (750nM, SelleckChem, S2858), and UM729 (500nM, STEMCELL, #72332).
Cord blood hematopoietic progenitor cells were isolated from cord blood samples using the EasySep™ human progenitor cell enrichment kit (STEMCELL, #19356) and cultured in StemSpan SFEM II medium (STEMCELL, #09655) supplemented with 20% BIT 9500 (STEMCELL, #09500), 1% Gluta-Plus, mSCF (100ng/mL), FLT3-L (100ng/mL), and TPO (100ng/mL).
All cell lines and human samples were maintained in an incubator at 37°C with 5% CO2.
Lentivirus production and transduction
HEK293T cells were grown in RPMI 1640 medium (Wisent, #350-035-CL) supplemented with 10% FBS (Wisent, #080–150) and 1% Gluta-Plus (Wisent, #609-066-EL). Cells were seeded in 15cm tissue culture plates (Sarstedt, #83.3903) at a density of 7 x106 cells per plate one day before transfection. On the day of transfection, cells were co-transfected with lentiviral plasmid vectors, psPAX2 (Addgene, #12259), and pCMV-VSVG (Cell biolabs, #320023) using the jetPRIME transfection reagent (Polyplus, #CA89129-924) according to manufacturer’s protocol. Supernatant containing viral particles was collected at 48- and 72-hours post transfection and filtered through a 0.45 µm PVDF membrane (Sigma, #SE1M003M00). Viral particles were precipitated in 40%(w/v) polyethylene glycol (Sigma, #89510-1KG-F) overnight. On the next day, the viral particles were collected by centrifugation at 3,700rpm for 30 mins at 4°C. The pellet was resuspended in HBSS (Gibco, #14170112) + 25mM HEPES (Thermo Fisher, #15630-080) and stored at -80°C for long term storage.
For lentiviral transductions, non-TC-treated 24 well plates were coated with 20 µg/mL of Retronectin (Takara, Cat # T100B) for 2 hours at room temperature followed by aspiration and blocking with PBS containing 2% (w/v) BSA (Wisent Bioproducts, Cat # 800-096-EG) for 30 mins at room temperature. After aspiration of the blocking buffer, the concentrated virus suspension was added to wells. The plates were then centrifuged at 3,700 rpm for two hours at 4°C to allow virus binding. Following centrifugation, unbound virus was aspirated, and cells were added. The plates were then transferred to a 37°C incubator to initiate lentiviral infection.
Generation of Venus‒ cells using CRISPR/Cas9
A synthetic sgRNA targeting Venus was ordered from Synthego. See Table S2 for sequence. Recombinant S.pyogenes Cas9 endonuclease at a concentration of 10ug/uL (62uM) was purchased (IDT, #1081058). The sgRNA was reconstituted in 15uL of TE buffer to reach a stock concentration of 0.1mM. 2.5uL of the sgRNA was then mixed with 1.5uL of SpCas9 in a final volume of 25uL of Lonza P3 nucleofection solution (Lonza, #V4XP-3024) and incubated at room temperature for 15 minutes to allow formation of the sgRNA/Cas9 RNP complex. 1x106 Tet2WTHPCHOXB4 cells were resuspended in 75uL of P3 nucleofection solution, added to the sgRNA/Cas9 RNP complex, and transferred to a Lonza nucleofection microcuvette. Nucleofection was conducted using a Lonza Amaxa™ 4D-Nucleofector under the program CA137. The presence of Venus knockout (Venus‒) cells was assessed by flow cytometry 4 days after nucleofection.
In vitro competition assays
Competition assays were conducted in 96-well round-bottom tissue culture plates (Sarstedt, #83.3925) with a total of 1×104 cells seeded in each well.
For competition assays involving HPCHOXB4 cells, BFP‒ or Venus+ HPCHOXB4 cells were mixed with BFP+ or Venus‒ competitor cells, respectively, at a 1:4 starting ratio in HPCHOXB4 growth medium to start the competition. BFP+ competitor cells were generated by transducing Tet2WTHPCHOXB4 cells with the pRSI9-U6-sh-UbiC-TagBFP-2A-puro vector (Addgene, #28289). Venus‒Tet2WT HPCHOXB4 competitor cells were generated as above. The BFP+ and Venus‒ competitor cells were sorted to > 99% purity using a Becton Dickinson Aria III CFI cell sorter.
For competition assays involving unmodified murine bone marrow HSPCs, CD45.2+ and CD45.1+ whole bone marrow cells were harvested from age-matched mice (10–15 weeks). Lin-Kit+ bone marrow HSPCs were enriched using the EasySep mouse hematopoietic progenitor isolation kit (STEMCELL, #19856) followed by the c-KIT positive enrichment kit (STEMCELL, #18757). Enriched CD45.2+ and CD45.1+ cells were then mixed at a 1:4 starting ratio and cultured in MethoCult™ GF M3434 medium (STEMCELL, #03434) supplemented with recombinant human TPO (25ng/mL).
Chemical library screen
Epigenetic probes were dissolved in DMSO to achieve a 20,000X stock solution and then diluted in PBS to achieve a 200X working stock solution. BFP+Tet2WTHPCHOXB4 was generated by lentiviral transduction of Tet2WTHPCHOXB4 with a pCDH-EF1-TagBFP-T2A vector and enriched by FACS sorting. BFP‒Tet2KOHPCHOXB4 cells were mixed with BFP+Tet2WTHPCHOXB4 competitor cells at the ratio of 1:4 and then seeded in 96-well round-bottom TC-treated plates at 1x104 total cells per well. The cells were cultured in HPCHOXB4 growth medium and treated with each epigenetic probe at a final concentration shown in Table S3 or DMSO (0.01%). Cells were split at 1:10 ratio on day 7 by adding 20uL of cells from old wells to 180uL of media containing epigenetic probes at screen concentration. The percentage of BFP+ cells in each well was measured by flow cytometry after 14 days of treatment.
Flow cytometry
For staining of cell surface antigens, cells were incubated with antibodies at the recommended dilutions (Table S4) in FACS buffer (HBSS supplemented with 2% FBS and 0.1% sodium azide) for 20 mins at 4°C. The cells were washed once in FACS buffer prior to analysis. For Annexin V staining, cells were resuspended in Annexin V buffer and stained with APC-conjugated Annexin V diluted in 1:200 (Invitrogen, #A35110) for 15 mins at room temperature prior to analysis. Flow cytometry analysis was conducted using the Beckman Coulter CytoFLEX analyzer. FCS files were analyzed using the FlowJo™ V10 software.
Intracellular flow cytometry
Intracellular flow cytometry staining for phospho-STAT5 in murine bone marrow cells was performed using the BD Cytofix/Cytoperm fixation and permeabilization kit according to the manufacturer’s protocol. Briefly, fixed and permeabilized cells were stained with a phospho-STAT5 antibody (CST, #9359) diluted 1:200 in HBSS supplemented with 2% FBS and 0.1% sodium azide for 1 hour at room temperature. The cells were then washed in FACS buffer and stained with an Alexa Fluor 647-conjugated anti-rabbit IgG (H + L) secondary antibody (CST, #4414S) diluted 1:2,000 in PBS with 1% FBS and 50mM EDTA for 30 mins at room temperature. A rabbit IgG isotype antibody (CST, #2985S) was used to determine the level of isotype control staining.
EdU proliferation assay
One day prior to the assay, drug-treated Tet2WT and Tet2KO HPCHOXB4 cells were seeded in wells in a 24-well tissue culture plate at a cell density of 1×105 cells/mL. On the next day, EdU dissolved in DMSO was directly added to the growth medium at a final concentration of 10uM. The cells were incubated with EdU for 1 hour at 37°C prior to staining. Staining for EdU incorporation was performed using the Click-iT Pacific Blue EdU flow cytometry assay kit (Invitrogen, #C10636) as per the manufacturer’s protocol.
Colony-forming unit (CFU) assays
CD34+ human HSPCs were enriched from cord blood samples using the CD34+ positive selection kit (STEMCELL #17856). 1x104 enriched Sca-1+ murine HSPCs or CD34+ human HSPCs were resuspended in 1.1mL of MethoCult™ GF3434 medium or MethoCult™ H4435 medium, respectively. The medium was supplemented with recombinant human TPO (25ng/mL). The resuspended cells were added to wells in a 6-well tissue culture plate. The number of colonies formed was determined after 7 days in culture and if necessary, the cells were replated.
RNA-sequencing analysis
Total RNA was extracted from cells using the Qiagen RNeasy plus kit (Qiagen, #74134) and quantified using a Nanodrop spectrophotometer. All samples had a RIN value greater than 9. Samples were submitted to Novogene Corporation for sequencing analysis. Library construction, sequencing, and processing of sequencing data were done by Novogene following their standard pipeline. Sequencing was performed on an Illumina NovaSeq 6000 system. Reads containing adaptors and low-quality reads with Phred scores less than 33 were filtered. Clean reads were then mapped to the reference genome using the STAR software v2.6.1d. Quantification of mapped reads was conducted on FeatureCounts (v1.5.0-p3) program and default parameters were applied.
For differential gene expression (DGE) analysis, triplicate pairwise samples were analyzed using DESeq2 R package (v1.20.0). P-values were adjusted using the Benjamini and Hochberg’s approach. |log2FC| > 1.5 and padj < 0.01 were used as the cutoffs for differentially expressed genes.
Chromatin-immunoprecipitation sequencing (ChIP-seq) analysis
Cells were fixed with 1% formaldehyde (Thermo Scientific, #410730050) according to the Active Motif ChIP cell fixation protocol. Fixed cell pellets were submitted to Active Motif for ChIP-seq analysis. Briefly, 15ug of chromatin and 10uL of H3K79me2 antibody (Active Motif, #39143) were used for each IP reaction. Illumina base-call data were processed and demultiplexed using bcl2fastq2 v2.20 and low-quality bases with Phred scores less than 33 were trimmed. 75 bp single-end sequence reads were subsequently mapped to the genome through BWA v0.7.12 algorithm with default settings. Low quality reads were filtered out and PCR duplicates were removed. Aligned sequencing reads, or tags, were extended to 200bp from the 3’ end, followed by dividing the genome into 32bp bins and counting the number of fragments in each bin. Determination of enriched regions was done using SICER 1.1 peak calling tool, with FDR < 1e-10 and gap parameter of 600bp used as cutoffs. False ChIP-seq peaks as defined within the ENCODE blacklist were removed. Differential methylation analysis was conducted using DiffBind R package version 3.10.0 and signals from each sample were normalized to library size. Hypermethylated regions in pairwise comparisons were defined using log2(FC in normalized reads) > 1.5 and padj < 0.01 as cutoffs. Hypomethylated regions were defined using log2(FC in normalized reads) < -1.5 and padj < 0.01 as cutoffs. Average peak signals from two replicates of each sample were obtained through randomly sampling 50 percent of reads from the corresponding BAM file of each replicate, and then merged using Samtools version 1.14. Average plots and heatmaps were generated using Deeptools version 3.5.1. BigWig files were visualized using WashU Epigenome browser.
RT-qPCR
Total RNA was extracted from cells using the Qiagen RNeasy plus kit and quantified on a Nanodrop spectrophotometer. Reverse transcription and quantitative PCR were performed using the Luna® Universal One-step RT-qPCR kit (NEB, #E3005S) and the Bio-Rad CFX touch real-time PCR detection system. The primers used are listed in Table S5. The threshold cycle (Ct) value was determined using CFX Manager v3.1. Gene expression was calculated using the ∆∆Ct method. Ct values were normalized to beta actin (Actb).
ChIP-qPCR
Lin-Kit+ bone marrow HSPCs were enriched from Tet2WT and Tet2KO mice using selection kits (STEMCELL, #19856 and #18757). Chromatin for each CHIP reaction was prepared with 106 HSPCs using Diagenode chromatin EasyShear kit following the manufacturer protocol (Diagenode # C01020010). Chromatin was sheared at 4ºC on a Bioruptor® Pico sonicator in 100uL volume with 30s on and 30s off for 20 pulses. Each ChIP reaction was conducted using 7uL of H3K79me2 antibody (Active motif, #39143) following a published protocol (Bailey SD et al., Nat Genet. 2016). DNA was purified using Zymo ChIP DNA clean and concentrator (Zymo research, #D5205) and eluted in 40uL of deionized water. qPCR was performed with Biobasic qPCR mastermix (Biobasic, #QPCR004-S) and the Bio-Rad CFX touch real-time PCR detection system. qPCR primers against Mpl and two negative control regions were ordered from IDT. Sequences of ChIP-qPCR primers are listed in Table S6. Enrichment was calculated using the percent input method.
Single-cell RNA (scRNA) sequencing
Whole bone marrow cells were isolated from 15 to 20-week-old Tet2WT and Tet2KO male littermates following red blood cell depletion. Cells from each donor were transplanted into four lethally irradiated 8-week-old CD45.2 Tet2WT recipients at 1x106 cells per animal. Following four weeks of engraftment, mice were subject to pinometostat (60mg/kg) or vehicle (10% DMSO, 40% PEG300, 5% Tween-80, and 45% saline) treatment through subcutaneous injection twice a day (2 animals per cell type per treatment group) for 21 days. Mice were then euthanized and Lin-Kit+ (LK) HSPCs were enriched from the bone marrow using selection kits (STEMCELL, #19856 and #18757). The purified cells were resuspended in IMDM medium (Wisent, #319-105-CL) and subjected to library preparation using 10x Genomics Chromium Single Cell 3′ v3.1 kit and 3’ sequencing on illumina Novaseq 6000 sequencing system. The FATSQ files were processed using Cellranger v7.0.0 pipeline to generate count matrices. Cells were filtered using the following cutoffs: 200 < nFeature < 9000; percent of mitochondrial reads < 12.5. The filtered reads were then normalized accounting for cell cycle difference using the SCTransform function from the Seurat package (v5.0.1). Samples showed a median of 22,504 Unique Molecular Identifiers (UMIs) per cell and 5,044 genes detected.
The filtered cells were subsequently assigned with a cell type label using the previously curated gene list (Izzo et al., Nat Genet. 2020). HSC = Hematopoietic stem cell; IMP = Immature myeloid progenitor, Mono = Monocyte progenitor, Neu = Neutrophil/granulocyte progenitor; EB = Erythroid/basophil progenitor; Ery = Erythroid progenitor; MkP = Megakaryocyte progenitor; CLP = Common lymphoid progenitor; Ba = Basophil progenitor; Eo = Eosinophil progenitor; B-cell-P = B cell progenitor; T-cell-P = T cell progenitor. Uniform Manifold Approximation and Projection (UMAP) reduction with the following parameters: reduction = “pca”, dims = 1:50 was employed for clustering.
AUCell (v1.25.0) package was used for gene signature enrichment analyses. Gene sets were imported from the molecular signatures database (MSigDB). Top 20 percent highest expressed genes were used in calculating the area under curve (AUC) for each gene set. Student T test was used to determine statistical significance between two groups.
Differential gene expression (DGE) analysis comparing Mpl+ vs Mpl- HSCs from the vehicle treated group was performed using FindMarkers() function of the Seurat package. Non-paramatric Wilcoxon rank sum test was used to calculate differentially expressed genes. |Log2FC| >1 was set as the fold-change cutoff. Only genes detected in equal or more than 0.01 percent of cells were tested. Genes passing the fold-change threshold were filtered again using adjust p value (padj) < 0.01 as cutoff. Cell type specific markers were imported from PanglaoDB (Franzén et al., Database. 2019) for Fig. 3I.
Western blot
Standard Western blotting techniques were performed. Blots were incubated with primary antibodies (Table S7) diluted in 5% (w/v) BSA in TBST overnight at 4°C on a shaking platform. On the next day, the blots were washed for five minutes for three times with TBST and incubated with a secondary antibody (LI-COR, #926-32213 or LICOR, #926-68073) at 1:2,000 dilution in 5% (w/v) BSA in TBST at room temperature for 1 hour. Membranes were washed three times again and imaged on the Odyssey CLx Imaging system (LI-COR Biosciences).
Droplet digital PCR (ddPCR)
Genomic DNA was extracted from treated cells. 125ng of DNA was used for each ddPCR reaction. The ddPCR reaction was prepared using the Bio-Rad ddPCR Supermix (Bio-Rad, #1863010). SNP assay primers and probes were ordered from Thermofisher (Table S8). Plates were read using a QX200 Droplet Digital PCR System (Bio-Rad). Results were analyzed using the QuantaSoft™ software.
In vivo experiments
All in vivo experiments were performed in accordance with institutional guidelines approved by the University Health Network animal care committee. Study design and data collection were in accordance with the ARRIVE guidelines.
For the experiment testing the effect of ruxolitinib, CD45.2+ Tet2KO BM cells from a 12-week-old male donor were mixed with CD45.1+ Tet2WT BM cells from age-matched male donors at a 3:7 ratio. The mixed cells were resuspended in Opti-MEM medium and transplanted by tail vein injection into 8-week-old female CD45.2+ Tet2WT recipient mice (1x106 cells per mouse) conditioned with 12Gy of irradiation. At day 21 post-transplantation, the mice were randomized to receive vehicle or ruxolitinib. Ruxolitinib (Table S9) was dissolved in DMSO and then diluted in peanut oil. Mice were administered 2% DMSO in peanut oil (vehicle control) or ruxolitinib (60mg/kg) by oral gavage twice a day.
For the experiment testing the effect of Mpl knockdown, lineage-depleted (Lin-) BM HSPCs from a 12-week-old male CD45.2+Tet2KO mouse were mixed with Lin- BM HSPCs from two age-matched CD45.1+Tet2WT male mice at a 3:7 ratio. The mixed cells were co-transduced with a pRSITE-U6-shMpl-UbiC-TagBFP-T2A-Puro or pRSITE-U6-shNT-UbiC-TagBFP-T2A-puro lentiviral vector (Table S10). BFP+ cells were sorted on day 3 after transduction, resuspended in Opti-MEM, and transplanted by tail vein injection into lethally irradiated 8-week-old Tet2WT female mice (1x104 cells per mouse). At day 21 post-transplantation, doxycycline (Table S9) was added into the drinking water at a concentration of 2mg/mL to induce shRNA expression and replaced once a week.
Peripheral blood chimerism analysis
Peripheral blood samples were collected and incubated in 1X red blood cell lysis buffer (Biolegend, #420302). The RBC-depleted cells were incubated in 50uL of PBS containing a FcR blocker (Biolegend, #426103) at 1:2,000 dilution for 10 min at 4°C and then stained with fluorophore-conjugated antibodies in FACS buffer for 20 min at 4°C. The stained cells were washed and resuspended in FACS buffer containing Sytox™ Green (Invitrogen, #S7020) at 1:1,000 dilution prior to flow cytometry analysis.
CRISPR/Cas9 mediated knockout of TET2 in human HSPCs
The synthetic TET2 KO-S sgRNAs (Table S2) were ordered from Synthego and resuspended in TE buffer to achieve a 100uM stock concentration. The synthetic TET2 KO-D and OR2W5P crRNAs were ordered from Integrated DNA Technologies and resuspended in TE buffer to achieve a 200uM stock concentration. TracrRNA was purchased from IDT (IDT, #1072532). To form the tracrRNA:crRNA complex, 1uL of each crRNA was mixed with 2uL of tracrRNA, heated at 95°C for 5 min on a thermocycler, and cooled. To form RNPs, 1.7uL of SpCas9 was diluted in 2.1uL of pre-warmed PBS and incubated with 1.2uL of the crRNA:tracrRNA complex or 2.5uL of sgRNAs at room temperature for 15 minutes. 1uL of Cas9 electroporation enhancer (IDT, #1075915) was added to the RNP complex solution. Cord blood HSPCs were enriched and cultured for 48 hours prior to nucleofection. After culturing, 5×105 HSPCs were resuspended in 100uL of Lonza P3 nucleofection solution containing supplements. The RNP complex was added to the cells. The cells were transferred to a Lonza nucleofection microcuvette and electroporated using program DZ100 on a Lonza Amaxa™ 4D-Nucleofector. Following electroporation, 1mL of culture medium was added to each microcuvette, and the cells were transferred to wells in a 24-well tissue culture plate. The culture medium was changed on the next day. Genomic DNA was extracted 1 day after nucleofection and subjected to PCR amplification using the primers listed in Table S11. PCR products were purified and submitted for Sanger sequencing. Estimation of knockout efficiency was determined using the ICE CRISPR Analysis Tool (Synthego).
Statistical analysis
Data are shown as means ± SD, unless otherwise indicated. Unpaired two-tailed Student’s T test was used to determine the level of significance between 2 sets of values. If needed, P-values were corrected for multiple comparisons using the Holm-Sidak method. P-values < 0.05 were considered significant. R bioinformatic analyses were conducted in R v4.3.2. Data were analyzed and visualized using the following R packages: ggplot2 (v3.4.4), ComplexHeatmap (v2.4.3), EnhancedVolcano (v1.6.0), VennDiagram (v1.7.3), ggpubr (v0.6.0), ggrepel (v0.9.4), AUCell (v1.24.0), msigdbr (v7.5.1), Seurat (v5.0.1), DESeq2 (v1.20.0).
Gene sets were downloaded from Molecular Signatures Database (MSigDB). Gene sets used for the enrichment analyses are: GRAHAM_NORMAL_QUIESCENT_VS_NORMAL_DIVIDING_UP; EPPERT_HSC_R; MALTA_CURATED_STEMNESS_MARKERS;PARK_HSC_VS_MULTIPOTENT_PROGENITORS_UP; IVANOVA_HEMATOPOIESIS_EARLY_PROGENITOR.