Hematopoietic stem cells (HSCs) have self-renewal capacity and multilineage developmental potentials. The molecular mechanisms that control the self-renewal of HSCs are still largely unknown. Here, a systematic approach using bioinformatics and array hybridization techniques to analyze gene expression profiles in HSCs is described. To enrich mRNAs predominantly expressed in uncommitted cell lineages, 54 000 cDNA clones generated from a highly enriched population of HSCs and a mixed population of stem and early multipotent progenitor (MPP) cells were arrayed on nylon membranes (macroarray or high-density array), and subtracted with cDNA probes derived from mature lineage cells including spleen, thymus, and bone marrow. Five thousand cDNA clones with very low hybridization signals were selected for sequencing and further analysis using microarrays on glass slides. Two populations of cells, HSCs and MPP cells, were compared for differential gene expression using microarray analysis. HSCs have the ability to self-renew, while MPP cells have lost the capacity for self-renewal. A large number of genes that were differentially expressed by enriched populations of HSCs and MPP cells were identified. These included transcription factors, signaling molecules, and previously unknown genes.

Hematopoiesis is a dynamic process with significant complexity in which a subset of hematopoietic stem cells (HSCs) gives rise to cells of both the myeloid and lymphoid lineages.1In addition, HSCs have the ability to self-renew to produce more HSCs. This property allows HSCs to repopulate the bone marrow of lethally irradiated congenic hosts.

Mouse HSCs isolated with Thy1.1loc-kit+Sca-1hiLin−/lomarkers using fluorescence activated cell sorting (FACS) represent only approximately 0.05% of mouse bone marrow cells and these cells can fully reconstitute all blood cell elements.2-4 This population of cells has been further divided into 3 subpopulations4,5: long-term HSCs (LT-HSCs), short-term HSCs (ST-HSCs), and multipotent progenitor (MPP) cells, according to their abilities to support hematopoiesis and self-renewal. These 3 populations of cells can be arranged in a lineage according to a progressive loss of the ability to self-renew.6 The LT-HSC population has extensive self-renewal ability and supports long-term reconstituting ability (> 6 months), representing approximately 0.005% to 0.01% of bone marrow cells.4 ST-HSCs with limited self-renewal ability briefly contribute to hematopoiesis (6-8 weeks). MPP cells cannot self-renew but can reconstitute bone marrow for less than 4 weeks.5 Efflux of dyes such as rhodamine-123 (Rh) can be used to separate early hematopoietic cells into HSC and early progenitor subpopulations by flow cytometry (see Pohlmann et al7 and Weissman8 for detailed description). Rh is a mitochondria-binding fluorescent dye, and can be effluxed from the cell by the ABC transporter including the multidrug-resistance (MDR) gene product, P glycoprotein.12LT-HSCs, which are relatively quiescent,13 have high ABC transporter activity, thus the population of cells that stain most weakly with this dye is highly enriched for LT-HSCs. Alternatively, LT-HSCs, ST-HSCs, and MPP cells can be identified according to the expression levels of lineage-associated antigens such as Mac-1 and CD4.4,5 

The rarity of the HSCs, coupled with an inability to maintain these cells in culture for a significant period, has greatly hindered the biochemical and molecular characterization of these cells. Although various growth factors can induce stem cells to proliferate in vitro, no known combination of growth factors has been shown to promote sustained self-renewal of the HSCs. Rather, stem cells induced to proliferate in vitro invariably undergo concomitant differentiation. HSC functions are thought to be regulated by interactions of stem cells with stromal cells.14 The dynamic process of HSC development, including self-renewal, expansion, and maturation, involves interactions between the intrinsic genetic program and extrinsic signals from stromal cells. This dynamic process is accompanied by global changes in gene expression profiles involving molecular events such as signal transduction, transcriptional and translational regulation, and chromatin modification. In the present study, we intend to systematically look into these molecular events by analyzing the gene expression profiles of self-renewing HSCs and non–self-renewing MPP cells. Therefore, any changes in the components of these aforementioned molecular events may provide insight into understanding the molecular mechanisms in controlling self-renewal of HSCs.

Isolation of HSCs and generation of cDNA libraries

For cDNA library A, 4.7 × 109 bone marrow cells were collected from the femurs and tibias of 60 C57BL/6J mice (6-8 weeks old). These bone marrow cells were incubated with rat monoclonal antibodies against lineage-positive markers (Pharmingen, San Jose, CA) including CD4 (RM4-4), CD2 (RM2-5), CD45R/B220 (RA3-6B2), Gr-1 (RB6-8C5), Mac-1 (M1/70), TER-119 (TER-119), and IL-7Rα chain (B12-1); lineage negative (Lin−/lo) cells were enriched by twice depleting lineage-positive cells through incubation with antibody-coated (sheep antirat IgG) Dynabeads M-450 (Dynal). The remaining Lin−/lo cells were stained with Sca-1 fluorescein isothiocyanate (FITC) for isolation of Lin−/loSca-1+ cells.

The cDNA library A was constructed from 1.3 × 106Lin−/loSca-1+ cells described above using the ZAP expression cDNA synthesis kit following the manufacturer's procedure (Stratagene). Briefly, total RNA was isolated from the Lin−/loSca-1+ cells using Trizol Reagent (BRL). The cDNA was synthesized first by reverse transcription using Superscript II (Invitrogen, Carlsbad, CA) and then by DNA synthesis using Klenow DNA polymerase (Invitrogen). The cDNA inserts were cut with EcoRI and XhoI restriction enzymes and cloned into the EcoRI/XhoI sites of λZAPExp vector. The plasmids (pBK; Stratagene, La Jolla, CA) bearing the cDNA inserts were excised from their parent vector λZAPExp using helper phage according to the manufacturer's protocol. The primary cDNA library contained 36 000 clones.

The cDNA library B was made from 2.8 × 104 twice-sorted Lin−/loSca-1+Thy-1.1loc-kit+HSCs. Isolation of a population of Lin−/loThy-1.1loc-kit+Sca-1+ HSCs, RNA extraction, cDNA synthesis, and polymerase chain reaction (PCR) amplification have been described previously.15 There were 7.5 × 106 clones in the original library.

Probe preparation for macroarray and microarray analyses

For preparation of cDNA probes used for macroarray hybridization, total RNA isolated from lineage-positive cells derived from thymus, spleen, and bone marrow was used as template to generate the first strand of cDNAs using SuperScript II (Invitrogen), incorporating 33P-dCTP into the cDNA probes during the reverse transcription.

For preparation of cDNA probes used for microarray hybridization, c-kit-phycoerythrin (PE) (2B8) and Sca-1-biotin (D7)/Streptavidin-Cy-Chrome (Pharmingen) were used to stain Lin−/lo cells prior to separation using a Vantage fluorescence-activated cell sorter (Becton Dickinson, San Jose, CA). In order to isolate HSCs from MPP cells, Rh (0.1 μg/mL; Sigma, St Louis, MO) was used to separate Lin−/loc-kit+Sca-1+ cells into Rhlo (lowest 15%) and Rhhi (highest 15%) populations. We were able to obtain 1.4 × 104RhloLin−/lo c-kit+Sca-1+and 1.5 × 104RhhiLin−/loc-kit+Sca-1+cells from 15 mice. The postsorting analysis showed that the purity of the sorted Lin−/loc-kit+Sca-1+cells was 98%. Total RNA was isolated from equal numbers of RhloLin−/lo c-kit+Sca-1+and RhhiLin−/loc-kit+Sca-1+cells (1 × 104). In order to amplify the cDNA for microarray hybridization, PCR amplification was performed using pSMART cDNA synthesis strategy following the manufacturer's procedure (ClonTech, San Jose, CA). Briefly, VNdT20(5′-GACTCTAGAGCGGCCGCCTTTTTTTTTTTTTTTTTTTTVN-3′, V = A, G, C; n = T, A, G, C) was used for the first-strand cDNA synthesis. 5′-cap oligo (5′-TACGGCTGCGAGAAGACGACAGAAGGG-3′) was used for the second-strand cDNA synthesis. Both the VNdT and 5′-cap primer (5′-TACGGCTGCGA GAAGACGACAGAA-3′) were used for the PCR amplification with limited amplification cycles (20 cycles were used in this study). Cy3-dCTP (for Rhlo cells) and Cy5-dCTP (for Rhhi cells) fluorescence were incorporated during the PCR reaction. The unincorporated nucleotides were removed through a Millipore column (Millipore, Bedford, MA). The optical density (OD)200-700nm of the probes was measured using a spectrophotometer. The incorporation rate was calculated subsequently according to the ratio of the absorbance of Cy3 (523 nm) and Cy5 (633 nm) over the DNA absorbance at 260 nm.

Macroarray preparation, hybridization, and analysis

To prepare for macroarray, the 2 HSC cDNA libraries, A and B, described above were plated onto LB agarose/ampicillin plates in 24 × 24-cm Q-trays (Genetix, Christchurch, England) with densities of 500 to 2000 colonies per tray, and incubated overnight at 37°C. The bacterial colonies were then picked up by a Q-bot robotic system (Genetix) and inoculated into 384-well plates. Thus, each cDNA clone was addressed by its position in the plate. The macroarrays were generated by spotting bacterial colonies from 384-well plates onto 24 × 24-cm nylon membranes using Q-bot (Genetix). Thirty-six thousand cDNA clones from library A and 18 000 clones from library B were spotted in duplicate, and the position of each clone on the membrane was recorded according to its address in the 384-well plate. These macroarray membranes were incubated on the top of LB-agarose plus antibiotics at 30°C overnight in Q-trays. DNAs were denatured by soaking the membrane in 0.5 N NaOH/1.5 M NaCl for 10 minutes and neutralized with 0.5 M Tris-Cl, pH 8.0/1.5 M NaCl for 5 minutes. DNAs were fixed by ultraviolet (UV) crosslinking (UVP, Upland, CA). For hybridization, membranes were prehybridized for 2 hours at 60°C in 100 mL solution (12.5 mM PEG8000; 250 mM NaCl; 85 mM Na2HPO4; 7.5 mM H3PO4[86%]; 243 mM sodium dodecyl sulfate [SDS]; 10 mM ethylenediaminetetraacetic acid [EDTA]; 32 mM NaOH, pH 7.2) containing 100 μg/mL of denatured salmon sperm DNA and 100 μg/mL denatured mouse Cot-1 DNA. Denatured 33P-labeled probes from mature cell lineages were added to the prehybridized solution and incubated overnight in roller bottle or shaking bath at 60°C. Membranes were washed twice with 2x sodium chloride sodium citrate (SSC)/1% SDS at room temperature for 15 minutes, and then once in 0.2x SSC/0.1% SDS at 60°C for 30 minutes. Membranes were exposed to a phosphor imager screen and scanned using a phosphoimage scanner (Molecular Dynamics, Sunnyvale, CA). The hybridization result was analyzed using Spotfinder for image processing (H. Hammersmark and R.B., University of Washington, Seattle). Since each cDNA clone was spotted in duplicate, an average signal intensity of the duplicate was used. Local background hybridization signals surrounding each spot were subtracted prior to comparing spot intensity. Quantitative comparison of signal intensities was analyzed using Microsoft Excel. In order to avoid false-negative signals generated from slow or nongrowing bacterial colonies, a probe derived from the vector sequence was hybridized to the macroarray in parallel. The negative selection process was performed by selecting cDNA clones with positive signals for vector hybridization and negative or very weak signals for the cDNA probes derived from lineage-positive cells. Using this process, 3000 of 36 000 cDNA clones and 2000 of 18 000 cDNA clones were selected from library A and B, respectively, for microarray analysis.

Microarray preparation, hybridization, and analysis

cDNA inserts from 5000 clones selected using macroarray analysis described above were PCR-amplified using primers derived from the vector. For library A, ZAP-F (5′-AGTGGATCCAAAGAATTC-3′) and ZAP-R (5′-CTCTAGAAGTACTCTCGAG-3′) were used, and for library B, SubA1 (5′-CTTCGAACCGCGGATATCAGATC-3′) and SubS2 (5′-AAGGTTCCTTCACAAAGATCCCTCGAG-3′) were used. Amplified PCR products were purified using Sephacryl S500 (Pharmacia, Peapack, NJ), mixed 1 to 1 with dimethyl sulfoxide (DMSO) (Amersham, Piscataway, NJ), and spotted in duplicate onto each 75 × 25-mm coated type 7 microarray slide (Amersham) using Genespoter II (Molecular Dynamics). The microarray slides were air-dried and DNA was fixed by UV crosslinking at 500 mJ. The microarray slides were hybridized with Cy3- and Cy5-labeled probes described above. Briefly, the purified probes were concentrated with a speed-vacuum in the dark and resuspended in 20 μL hybridization buffer (5x SSC, 5x Denhardt, 0.1% SDS, 50% formamide, 100 μg/mL denatured salmon sperm DNA, 20 μg/mL polyA60 RNA, 100 μg/mL mouse Cot I DNA). An equal amount of Cy3- and Cy5-labeled probes were combined, denatured, and added onto the microarray slides, and hybridized at 42°C in a hybridization oven or in a humidified chamber for 16 hours to 18 hours. Microarray glass slides were washed twice with 2x SSC/0.1% SDS, and 0.2x SSC/0.1% SDS at 55°C for 5 minutes, and then with 0.1x SSC at room temperature for 1 minute. Glass slides were immersed in water for 10 seconds and dried immediately with N2. The hybridized microarray was scanned with a confocal dual-laser scanner at 523 nm and 633 nm (Molecular Dynamics) and analyzed using the custom array analysis software developed at the University of Washington (H. Hammersmark and R.B.). This array analysis software includes image processing, data normalization, and error analysis as described previously.16 

Reverse transcriptase–polymerase chain reaction

Reverse-transcription reactions were carried out using SuperScriptII following the manufacturer's manual (Invitrogen). The following primers were used for PCR: Activin βC, 5′-GACACCTTACTCTGGAGCTG-3′ and 5′-GGGAGGCAGA GTAGATTACA-3′; BA_RA6B66, 5′-TCATGGTATGCCCTCGTGTA-3′ and 5′-AAATGTGTGGGCTTTTCAGG-3′; BA_RA5A82, 5′-AACATGGCTTGGAGAC AACC-3′ and 5′-AAGCCTGGGATTCACTGCTA-3′; BA_RA4B58, 5′-ATGAGGGC CATTGTTACACC-3′ and 5′-TTATGGCCAGCTTGGTTCAC-3′; MSC_RA15C76, 5′-TGATACCCTTGGCTCGAAAC-3′ and 5′-CCAAGTGCTGGGATTAAAGG-3′; MSC_RA16D13, 5′-GGCTCGAAATTAACCCTCAC-3′ and 5′-CCAGATCTCGT TACGGATGG-3′. The PCR condition was 35 cycles consisting of 94°C for 30 seconds, 55°C for 40 seconds, and 72°C for 1 minute.

Competitive repopulation assay

Two hundred fifty cells (either RhloLin−/loSca-1+c-kit+or RhhiLin−/loSca-1+c-kit+) from Ly5.2 mice were mixed with 4 × 104Lin−/lo cells from congenic host Ly5.1 mice17and were then transplanted into lethally irradiated (11 Gy [1100 rads]) Ly5.1 mice by intravenous (IV) injection. Each group included 8 mice. Peripheral blood cells were harvested at different time points after transplantation, as indicated in Figure 2C, and flow cytometric analyses were performed using different specific cell surface markers (CD3, B220, Mac-1, and Gr-1) together with anti-Ly5.2 antibody.

Construction and normalization of HSC cDNA libraries

In order to enrich for genes that are predominantly expressed in the uncommitted, or lineage-negative cell populations, and remove highly redundant and housekeeping genes for further analysis, high-density array18 (macroarray) analysis was used and cDNA clones that hybridized to the lineage-positive probes were removed. cDNA clones that had no or very weak hybridization signals to the lineage-positive probes were selected. These negatively selected cDNA clones were subsequently subject to analysis of differential gene expression using a microarray system.19 The strategy for using macroarrays and microarrays to analyze the gene expression patterns of the HSC is illustrated and described in Figure1A. First, 2 populations of HSCs were isolated from mouse bone marrow cells by FACS based on their cell surface antigen expression: the Lin−/loSca-1+population (A) containing MPP cells, LT-HSCs, and ST-HSCs20; and the Lin−/loc-kit+Thy-1loSca-1+population (B) enriched with LT-HSCs.5 Correspondingly, cDNA libraries A and B were generated from each of these 2 complementary populations of HSCs. Sequence analysis of 200 randomly chosen clones from each library indicated that they are very diverse. Bacterial colonies derived from these HSC cDNA libraries were arrayed onto nylon membranes using the Q-bot robotic system to generate the macroarrays, or high-density cDNA arrays. These macroarrays were hybridized with the cDNA probes generated from committed or mature hematopoietic cell lineages of the thymus, bone marrow, and spleen (lineage-positive probes). Clones that hybridized to the lineage-positive probes were eliminated through the process of negative selection (see “Materials and methods”) and the remaining clones were selected and spotted onto microarray type glass slides. These microarrays were used to analyze the gene expression profiles of HSCs and MPP cells by hybridizing with fluorescent-labeled (Cy3 and Cy5) cDNA probes derived from the sorted cell populations of HSCs and MPP cells. This allowed the identification of genes preferentially expressed in the HSCs and MPP cells, respectively.

Fig. 1.

Enrichment of genes that are predominantly expressed in hematopoietic stem cells.

(A) A schematic drawing illustrating the strategy of systematic analyses of gene expression in hematopoietic stem cells (HSCs). Bacterial colonies from the 2 HSC cDNA libraries A and B described in the text were spotted in duplicates onto 24 × 24 cm nylon membranes using a Q-bot robotic system (Genetix), generating macroarrays. The macroarray membranes were hybridized with 33P-labeled cDNA probes derived from mature cell lineages described in the text in parallel with the vector-derived probe. Hybridization signals were analyzed using Spotfinder software (see “Materials and methods” for detailed description) and converted to numeric numbers using Microsoft Excel. cDNA clones, showing negative or very weak hybridization signals to the cDNA probes derived from lineage-positive cells but positive to the vector-derived probe, were selected. This process was called negative selection. The negatively selected cDNA clones were analyzed by DNA sequencing, and spotted in duplicate onto microarray slides. Total RNA isolated from RhloLin−/loc-kit+Sca-1+or RhhiLin−/loc-kit+Sca-1+HSCs was used to generate Cy3- and Cy5-labeled cDNA probes, respectively, by RT-PCR. The probes were hybridized to the microarray slides and the data were analyzed using array analysis software. Details of the experiment are described in “Materials and methods.” (B) Northern analysis of unknown genes from cDNA library B. Two micrograms of poly A+ RNA purified from indicated tissues was separated on a 1% agarose-formaldehyde gel, transferred to nylon membrane, and then hybridized with a 32P-labeled probe prepared from each unknown clone. Four representative blots are shown.

Fig. 1.

Enrichment of genes that are predominantly expressed in hematopoietic stem cells.

(A) A schematic drawing illustrating the strategy of systematic analyses of gene expression in hematopoietic stem cells (HSCs). Bacterial colonies from the 2 HSC cDNA libraries A and B described in the text were spotted in duplicates onto 24 × 24 cm nylon membranes using a Q-bot robotic system (Genetix), generating macroarrays. The macroarray membranes were hybridized with 33P-labeled cDNA probes derived from mature cell lineages described in the text in parallel with the vector-derived probe. Hybridization signals were analyzed using Spotfinder software (see “Materials and methods” for detailed description) and converted to numeric numbers using Microsoft Excel. cDNA clones, showing negative or very weak hybridization signals to the cDNA probes derived from lineage-positive cells but positive to the vector-derived probe, were selected. This process was called negative selection. The negatively selected cDNA clones were analyzed by DNA sequencing, and spotted in duplicate onto microarray slides. Total RNA isolated from RhloLin−/loc-kit+Sca-1+or RhhiLin−/loc-kit+Sca-1+HSCs was used to generate Cy3- and Cy5-labeled cDNA probes, respectively, by RT-PCR. The probes were hybridized to the microarray slides and the data were analyzed using array analysis software. Details of the experiment are described in “Materials and methods.” (B) Northern analysis of unknown genes from cDNA library B. Two micrograms of poly A+ RNA purified from indicated tissues was separated on a 1% agarose-formaldehyde gel, transferred to nylon membrane, and then hybridized with a 32P-labeled probe prepared from each unknown clone. Four representative blots are shown.

Close modal

To generate the macroarrays of cDNA libraries A and B, 36 000 clones from library A (made from Lin−/loSca-1+ cells) and 18 000 of 7.5 × 106 cDNA clones from library B (made from Lin−/lo c-kit+Thy-1loSca-1+cells) were arrayed onto nylon membranes (Figure 1), and hybridized with 33P-labeled cDNA probes derived from the hematopoietic lineage-positive populations of cells. The image data were analyzed using Spot-Finding software,16 which converted the image intensity of each spot into numerical data. After analyses, 3000 cDNA clones from macroarray A and 2000 clones from macroarray B that were negative or weakly positive were chosen to further construct the microarray. We have analyzed these negatively selected cDNA clones. A sequence comparison of 384 randomly chosen clones after the negative selection indicated that: (1) the percentage of housekeeping gene sequences represented by sequences encoding for mitochondrial and ribosomal proteins was greatly reduced from 25% to 5%; (2) the percentage of unique genes was increased to 60%; and (3) the percentage of novel sequences, which have no significant homologies with known sequences, was increased to 10% at the time the present manuscript was submitted, albeit the percentage decreased with the rapid progress of human and mouse genomic projects (data not shown).

The sequence analysis of the 2000 negatively selected clones from library B provides a glimpse of the gene expression profile of adult bone marrow enriched with HSCs. A partial annotated list of genes identified in cDNA library B is shown in Table1. Numerous gene encoding transcription factors, chromatin modifiers, transmembrane proteins, and signaling molecules were identified. Based on their molecular and cellular roles in other tissues, contexts, or species, these genes might be likely candidates for playing an essential role in stem cell functions. For example, the bmi-1 gene is a Polycomb-group gene that represses expression of Hox genes during development, and regulates cell proliferation and senescence by down-regulating theink4a locus.21 The ink4a locus expresses 2 genes by alternative splicing: p16 is a G1 cyclin inhibitor and p19ARF stabilizes expression of p53 by binding to Mdm2. Constitutive expression of both bmi-1 and telomerase results in immortalization of epithelial cells.22 Telomerase is activated in most immortal cell lines and cancers, and possibly is involved in the regulation of HSC self-renewal.23 The Notch family has been implicated in the determination of HSC fate.24-26 Recently, constitutive expression of the activated form of Notch1 was shown to be able to immortalize pluripotent, cytokine-dependent HSCs.27 

Table 1.

A partial list of genes analyzed after negative selection of the cDNA library generated from Lin−/loSca-1+Thy-1.1loc-kit+cells

NameClone codeAccession numberDescription
Transcription factors    
 Bmi-1 BA_HSC3_18 M64279 Polycomb-group transcriptional repressor; involved in cell proliferation and senescence  
 Nfe2 BA_RA1B_64 L09600 AP-1–related DNA-binding protein, leucine zipper protein, hematopoietic specific 
 TBX2 BA_RA3A_94 AF172088 Limb morphogenesis; immortalizing gene with tumorigenic potential 
 Ash1 BA_RA2A_52 NM_018489 A SET domain, a PHD finger, 4 AT hooks, and a region with homology to the bromodomain; may be involved in adhesion-mediated transcription 
 Maid BA_RA2B_83 NM_010761 Negative regulator of bHLH proteins in the mouse egg and zygote 
 LEF-1S BA_RA5D_78 D16503 A lymphoid-specific protein with an HMG domain, regulates TCRα enhancer function 
 Elf1 BA_RA6B_30 NM_007920 Ets-family transcription factor; highest in lymphocytes 
 ZF216 BA_RA4C_60 AF062071 1 N-terminal A20 (an inhibitor of cell death)-like zinc fingers, mediating IL-1–induced NF-kappaB activation  
 Evi-1 BA_RA3A_50 M21829 2 sets of ZF; overexpression causes abnormalities in megakaryopoiesis 
 Evi-9 BA_RA1D_75 NM_016707 A ZF protein; interacts with BCL6, a known human B-cell proto-oncogene product 
 Tcf4 BA_RA1A_07 NM_013685 A splicing variant of the ITF-2 transcript encoding a transcription factor that inhibits MyoD activity  
 Chx10 BA_RA2B_69 NM_007701 C elegans ceh-10 homeo-domain–containing homologue; developmentally regulated 
 Putative myelin regulatory factor 1 BA_RA4A_40 U14648 A brain-derived DNA-binding protein; regulates expression of myelin basic protein gene  
 KIAA0697 BA_RA4B_88 AB014597 1 TM and sequence-specific RNA binding by a novel KH domain; implicated in paraneoplastic disease and the fragile X syndrome  
Membrane proteins    
 Notch1 BA_8-3 NM_008714 Controls cell fate choices by inhibiting certain differentiation pathways 
 Type III multi-pass TM protein BA_RA1A_50 AF082535 4 TM; a gene similar to Homo sapiens GMP-17/NKG7/GIG-1; expressed by NK, CD8+, activated T, and macrophage R2 cells  
 Mouse homologue of Nessy BA_RA1A_60 NM_005768 Putative transmembrane protein Nessy, an evolutionarily conserved gene controlled by Hox proteins during Drosophila embryogenesis 
 CD34 BA_RA1C_19 S69295 Hematopoietic stem cell antigen, present in embryonic brain and adult brain, lung, and skeletal muscle 
 CGI-69 BA_RA4D_91 AF151827 Similar to human CGI-69 protein  
 HSA-A BA_RA4D_26 X56469 A mouse hematopoietic differentiation marker from B and T cells; signal peptide, 1 C-terminal TM  
 Human sperm surface Ag BA_RA4B_33 NM_003971 A novel human testis mRNA specifically expressed in testicular haploid germ cells; carrying leucine zipper dimerization motif  
Secreted molecules    
 BMP4 BA_RA6B_87 S65032 A signal peptide; secreted protein; TGF-beta family; exhibits mesoderm-inducing activity 
 L2G25B BA_RA4B_55 J04491 Signal peptide, CXC chemokine 
 SDF4 BA_RA5B_24 D50461 A signal peptide with Ca2+ binding motif  
 Flt3 ligand isoform E6 Δ16 BA_RA4A_50 S76464 Signal peptide; flt3 ligand isoform E6 Δ16, missing 1 TM, soluble form 
Apoptosis    
 Mcl 1 BA_RA3A_85 NM_008562 Myeloid leukemia sequence 1; a bcl-2–related gene; expressed in the early stage of embryonic stem cells  
 Heterochromatin protein 1α BA_RA2A_37 AF216290 Apoptosis-associated proteins in a human Burkitt lymphoma cell line  
Signaling molecules    
 WNK1 BA_RA1A_15 AF227741 9 TM, 3 coil-coil structures; similar to MEKK; unknown function 
 ARK2 BA_RA1A_91 U69107 Aurora-related kinase 2; involved in chromosomal segregation; expressed during S and G2/M phases; localized to the midbody 
 STK-1 BA_RA1A_91 D21099 Expressed in a large number of proliferating cells, but not in other tissues; Xenopus lavas XLP46 protein kinase; involved in cell growth 
 LynB Ba_RA1D_84 M57697 Tyrosine kinase; expressed in myeloid and B-lymphoid lineage cells  
 Truncated form of c-kit BA_RA6B_92 X65997 Kinase domain 
 GAP BA_RA5B_81 L13151 GTPase activating protein; inhibits ras-mediated signaling 
 RGS2 BA_RA6B_56 NM_00906 RGS box; GAP for trimeric G-protein; inhibits signaling from Gq 
 Nbr1 BA_RA1B_56 AF008676 B box and zinc finger domain–containing protein, unknown function; next to BRCA1gene  
 DMX-like 1 BA_RA6A_84 NM_005509 10 WD and 2 TM; highly conserved throughout revolution; involved in regulatory function 
 Sop2p-like BA_RA3B_29 Y08999 6 WD40; interacts with Arp3p and modulates profilin function  
Cell cycle    
 Cyclin H BA_RA4D_55 AF154914 Associates with cyclin-dependent kinase 7 and Mat1 to form the mammalian Cdk-activating kinase, CAK; suppressed by p53  
 Clk4 BA_RA1C_80 NM_007714 CDK-like kinase 4; phosphorylate spliceosome  
 PCM-1 BA_RA5A_71 L27841 6 coils; a 228-kd centrosome autoantigen with a distinct cell cycle distribution; from fetal liver 
 PACT BA_RA5B_02 U28789 p53 and pRB binding protein; binding to p53 and inhibits p53 DNA binding  
 Tousled-like kinase BA_RA4C_16 NM_011903 S/T kinase; 3 coils, NLS; cell cycle–dependent activity with highest activity during S phase and is regulated by phosphorylation 
Miscellaneous    
 p13MTCP1 BA_RA4D_10 U32332 Found in t(X;14) translocations  
 Meis 1 BA_RA6D_67 NM_010789 Myeloid ecotropic viral integration site 1; a PBX1-related homeobox gene involved in myeloid leukemia 
 RA-inducible protein E3 BA_RA6D_70 U29539 Present in the myeloid, B-lymphoid, and erythroid lineages, absent in nonhematopoietic cells  
 DJ-1 BA_RA2A_33 AB015652 A novel oncogene that transforms mouse NIH3T3 cells in cooperation with ras 
 HMG-1–related protein BA_RA3D_37 S50213 DNA-binding components of the V-(D)-J recombinase 
 LSP1 BA_RA6D_01 S74179 Lymphocyte-specific protein; binds F-actin; a negative regulator of neutrophil chemotaxis 
 Mea BA_RA2A_25 M27938 Male-enhanced antigen; involved in spermatogenesis 
NameClone codeAccession numberDescription
Transcription factors    
 Bmi-1 BA_HSC3_18 M64279 Polycomb-group transcriptional repressor; involved in cell proliferation and senescence  
 Nfe2 BA_RA1B_64 L09600 AP-1–related DNA-binding protein, leucine zipper protein, hematopoietic specific 
 TBX2 BA_RA3A_94 AF172088 Limb morphogenesis; immortalizing gene with tumorigenic potential 
 Ash1 BA_RA2A_52 NM_018489 A SET domain, a PHD finger, 4 AT hooks, and a region with homology to the bromodomain; may be involved in adhesion-mediated transcription 
 Maid BA_RA2B_83 NM_010761 Negative regulator of bHLH proteins in the mouse egg and zygote 
 LEF-1S BA_RA5D_78 D16503 A lymphoid-specific protein with an HMG domain, regulates TCRα enhancer function 
 Elf1 BA_RA6B_30 NM_007920 Ets-family transcription factor; highest in lymphocytes 
 ZF216 BA_RA4C_60 AF062071 1 N-terminal A20 (an inhibitor of cell death)-like zinc fingers, mediating IL-1–induced NF-kappaB activation  
 Evi-1 BA_RA3A_50 M21829 2 sets of ZF; overexpression causes abnormalities in megakaryopoiesis 
 Evi-9 BA_RA1D_75 NM_016707 A ZF protein; interacts with BCL6, a known human B-cell proto-oncogene product 
 Tcf4 BA_RA1A_07 NM_013685 A splicing variant of the ITF-2 transcript encoding a transcription factor that inhibits MyoD activity  
 Chx10 BA_RA2B_69 NM_007701 C elegans ceh-10 homeo-domain–containing homologue; developmentally regulated 
 Putative myelin regulatory factor 1 BA_RA4A_40 U14648 A brain-derived DNA-binding protein; regulates expression of myelin basic protein gene  
 KIAA0697 BA_RA4B_88 AB014597 1 TM and sequence-specific RNA binding by a novel KH domain; implicated in paraneoplastic disease and the fragile X syndrome  
Membrane proteins    
 Notch1 BA_8-3 NM_008714 Controls cell fate choices by inhibiting certain differentiation pathways 
 Type III multi-pass TM protein BA_RA1A_50 AF082535 4 TM; a gene similar to Homo sapiens GMP-17/NKG7/GIG-1; expressed by NK, CD8+, activated T, and macrophage R2 cells  
 Mouse homologue of Nessy BA_RA1A_60 NM_005768 Putative transmembrane protein Nessy, an evolutionarily conserved gene controlled by Hox proteins during Drosophila embryogenesis 
 CD34 BA_RA1C_19 S69295 Hematopoietic stem cell antigen, present in embryonic brain and adult brain, lung, and skeletal muscle 
 CGI-69 BA_RA4D_91 AF151827 Similar to human CGI-69 protein  
 HSA-A BA_RA4D_26 X56469 A mouse hematopoietic differentiation marker from B and T cells; signal peptide, 1 C-terminal TM  
 Human sperm surface Ag BA_RA4B_33 NM_003971 A novel human testis mRNA specifically expressed in testicular haploid germ cells; carrying leucine zipper dimerization motif  
Secreted molecules    
 BMP4 BA_RA6B_87 S65032 A signal peptide; secreted protein; TGF-beta family; exhibits mesoderm-inducing activity 
 L2G25B BA_RA4B_55 J04491 Signal peptide, CXC chemokine 
 SDF4 BA_RA5B_24 D50461 A signal peptide with Ca2+ binding motif  
 Flt3 ligand isoform E6 Δ16 BA_RA4A_50 S76464 Signal peptide; flt3 ligand isoform E6 Δ16, missing 1 TM, soluble form 
Apoptosis    
 Mcl 1 BA_RA3A_85 NM_008562 Myeloid leukemia sequence 1; a bcl-2–related gene; expressed in the early stage of embryonic stem cells  
 Heterochromatin protein 1α BA_RA2A_37 AF216290 Apoptosis-associated proteins in a human Burkitt lymphoma cell line  
Signaling molecules    
 WNK1 BA_RA1A_15 AF227741 9 TM, 3 coil-coil structures; similar to MEKK; unknown function 
 ARK2 BA_RA1A_91 U69107 Aurora-related kinase 2; involved in chromosomal segregation; expressed during S and G2/M phases; localized to the midbody 
 STK-1 BA_RA1A_91 D21099 Expressed in a large number of proliferating cells, but not in other tissues; Xenopus lavas XLP46 protein kinase; involved in cell growth 
 LynB Ba_RA1D_84 M57697 Tyrosine kinase; expressed in myeloid and B-lymphoid lineage cells  
 Truncated form of c-kit BA_RA6B_92 X65997 Kinase domain 
 GAP BA_RA5B_81 L13151 GTPase activating protein; inhibits ras-mediated signaling 
 RGS2 BA_RA6B_56 NM_00906 RGS box; GAP for trimeric G-protein; inhibits signaling from Gq 
 Nbr1 BA_RA1B_56 AF008676 B box and zinc finger domain–containing protein, unknown function; next to BRCA1gene  
 DMX-like 1 BA_RA6A_84 NM_005509 10 WD and 2 TM; highly conserved throughout revolution; involved in regulatory function 
 Sop2p-like BA_RA3B_29 Y08999 6 WD40; interacts with Arp3p and modulates profilin function  
Cell cycle    
 Cyclin H BA_RA4D_55 AF154914 Associates with cyclin-dependent kinase 7 and Mat1 to form the mammalian Cdk-activating kinase, CAK; suppressed by p53  
 Clk4 BA_RA1C_80 NM_007714 CDK-like kinase 4; phosphorylate spliceosome  
 PCM-1 BA_RA5A_71 L27841 6 coils; a 228-kd centrosome autoantigen with a distinct cell cycle distribution; from fetal liver 
 PACT BA_RA5B_02 U28789 p53 and pRB binding protein; binding to p53 and inhibits p53 DNA binding  
 Tousled-like kinase BA_RA4C_16 NM_011903 S/T kinase; 3 coils, NLS; cell cycle–dependent activity with highest activity during S phase and is regulated by phosphorylation 
Miscellaneous    
 p13MTCP1 BA_RA4D_10 U32332 Found in t(X;14) translocations  
 Meis 1 BA_RA6D_67 NM_010789 Myeloid ecotropic viral integration site 1; a PBX1-related homeobox gene involved in myeloid leukemia 
 RA-inducible protein E3 BA_RA6D_70 U29539 Present in the myeloid, B-lymphoid, and erythroid lineages, absent in nonhematopoietic cells  
 DJ-1 BA_RA2A_33 AB015652 A novel oncogene that transforms mouse NIH3T3 cells in cooperation with ras 
 HMG-1–related protein BA_RA3D_37 S50213 DNA-binding components of the V-(D)-J recombinase 
 LSP1 BA_RA6D_01 S74179 Lymphocyte-specific protein; binds F-actin; a negative regulator of neutrophil chemotaxis 
 Mea BA_RA2A_25 M27938 Male-enhanced antigen; involved in spermatogenesis 

In clone codes, RA refers to the clones that have been negatively selected by subtraction with mature hematopoietic cells. Note that Bmi-1 and Notch 1 were identified during the sequencing of the random clones. ZF indicates zinc finger; TM, transmembrane; KH, K homology RNA-binding domain; HMG, high-mobility group; TCR, T-cell receptor; RRM, RNA recognition motif.

The EST (expressed sequence tag) sequences that are predominantly expressed in hematopoietic stem cells are available at the web-accessible database at www.systemsbiology.org/research/stemcells. This EST database also applies to Tables 2 and 3.

HSCs express genes that are shared by other tissues such as brain, muscle, and germ line cells (Table 1). For example, male-enhanced antigen (Mea) specifically expressed in the testis is involved in spermatogenesis.28 Some of the genes we identified are specific to hematopoietic tissues; for example, RGS18 is expressed in LT-HSCs and ST-HSCs as well as monocytes.15 Among unknown genes, there are genes that code for a variety of important functional domains and several genes with no obvious homology to any known functional domains. Some of the other unknown genes were analyzed by Northern hybridization. Of 10 unknown clones tested, 2 were expressed in all tissues (no. 1), 2 were expressed in some tissues (no. 26), and 6 had no detectable signals in any of the tissues examined (no. 8, no. 11). These 6 clones are candidates for HSC-specific genes (Figure 1B).

Previously, Philips and his coworkers described the gene expression profile of Sca-1+AA4.1+c-kit+Lin−/locells isolated from fetal liver HSCs.29 There are a number of genes commonly expressed in both the adult bone marrow HSCs (Lin−/loc-kit+ Thy-1loSca-1hi) generated by this study and the fetal liver HSCs (Sca-1+AA4.1+c-kit+Lin−/lo) generated by Philips et al,29 such as Evi-1, TSA-1, Ramp1, RGS2, ZF216, macroH2A1.2, CD34, Notch1, vascular endothelial ZF1, and calmodulin. (Tables1-3). In comparison to gene expression profiles of neural progenitor cells,30 a number of genes were also found to be expressed in both neural progenitor and hemaptopoietic stem cells. These included histone H2A, interferon-induced factors, guanine triphosphate (GTP)–nucleotide binding protein, nuclear poly(a)-binding protein, GCN5 histone acetyltransferase, and so on, indicating a shared, at least partially, genetic program among HSCs and neural progenitor cells.

Table 2.

List of differentially expressed genes between RhloLin−/loSca-1+c-kit+and RhhiLin−/loSca-1+c-kit+HSCs

NameClone codeAccession numberBackgroundRhloRhhiLibrary
Transcription factor    Fold change Fold change  
 Nfix BA-RA6D21 NM_010906 Nuclear factor I family; role in development 3.5  
 Hoxa9 BA-RA1C45 M28449 Homologous to the Drosophila Hoxa9  2.4 B  
 Zfp207 BA-RA1D05 NM_011751 Zinc finger protein Zep; 2 C2H2 ZF, NLS 2.1  
 Enx-1 BA-RA1C35 NM_007971 Polycomb family, regulate Hox gene expression  2.5 
Kinase       
 PI3 kinase BA-RA5A82 BG354681 Phosphoinositide 3-kinase  
 PKA-like BA-RA4C80 BG354682 cAMP-dependent protein kinase 3.1  
Phosphatase       
 Tyr phosphatase BA-RA1D25 X63440 p19 protein-tyrosine-phosphatase  2.3 B  
RNA binding       
 RBM3 MSC-RA1A25 NM_016809 RNA binding motif 3 4.1  
 Napor-3 BA-RA6B66 AF090697 Apoptosis-related RNA-binding protein 2.7  B  
 Poly(a) binding BA-RA6B35 X65553 RNA-binding protein 2.3  
Protease       
 Calpain 4 MSC-RA17B72 NM_009795 Calcium-dependent protease 6.2  A  
Cell growth       
 Activin beta C MSC-RA15B58 X82540 TGF-beta family  4.3 A  
 TAX responsive MSC-RA15B86 X81987 Malignancy-related protein 107  3.5 A  
 Inhibitor of CDC42 MSC-RA16A75 L07918 Hematopoietic-specific, GDP dissociation inhibitor  21 A  
Protein modification       
 CCT eta MSC-RA12A61 Z31399 Chaperonin containing TCP-1 4.7  
 NEDD-4 MSC-RA16D13 P46935 Containing WW domain 2.1  A  
Membrane associated       
 pRGR1 BA-RA6C77 AF041429 4 TM; unknown function  3.9 
 TM-protein BA-RA4C93 AF082835 Type III transmembrane protein  3.7 B  
 Ly-6C.2 MSC-RA11C15 M18466 u-PAR domain proteins 6.1  A  
Nuclear protein       
 PWP1-like BA-RA5C56 L07758 WD-40, nuclear phosphoprotein  2.3 
 Npm1 BA-RA4A70 NM_008722 Nucleophosmin 1–related protein  
 Karyopherin BA-RA1C82 NM_010655 Importin  5.9 
Calcium binding       
 PW29 BA-RA4C42 D49429 Calcium-binding protein  3.9 B  
 Cam I BA-RA6C79 M19381 Calmodulin-dependent kinase and phosphatase  5.9 B  
Chromosome modeling       
 HAT1 BA-RA6D08 NM_003642 Histone acetyltransferase 1  A/B  
 Histone H2A BA-RA5A12 AF171080 Chromatin structure 3.1  A/B 
 Set beta isoform MSC-RA15B10 S68987 Inhibitor of HAT and PP2a  3.7 
 HMG1-related MSC-RA17A09 U00431 Chromatin organization  A/B 
Miscellaneous       
 Ant2 BA-RA2D75 AF240003 Adenine nucleotide translocase 2  4.1 
 Prg2 MSC-RA14C84 NM_008920 Major basic protein in eosinophils of bone marrow  4.9 A  
 Calponin 2 MSC-RA16D77 NM_007725 Actin binding protein  6.5 
 Transport-like proteins BA-RA1D09 AY008297 RPGR-interacting protein  A/B 
 CYLN2 BA-RA4A42 AAF99333 Cytoplasmic linker proteins  
Novel       
 KIAA0780 BA-RA1D26 BG354685 Unknown function  2.4 B  
 RP42 BA-RA2C82 BG354683 Unknown function  B  
 PTD011 BA-RA5D01 BG354684 Unknown function 2.9  
 Unknown BA-RA3D39 BG354687 Unknown function  2.1 
 Unknown MSC-RA16B18 BG354690 Unknown function 5.1  
 Unknown BA-RA4A52 BG354679 Unknown function  2.3 
 Unknown BA-RA4B58 BG354678 Unknown function  
 Unknown MSC-RA15C76 BG354689 Unknown function  
NameClone codeAccession numberBackgroundRhloRhhiLibrary
Transcription factor    Fold change Fold change  
 Nfix BA-RA6D21 NM_010906 Nuclear factor I family; role in development 3.5  
 Hoxa9 BA-RA1C45 M28449 Homologous to the Drosophila Hoxa9  2.4 B  
 Zfp207 BA-RA1D05 NM_011751 Zinc finger protein Zep; 2 C2H2 ZF, NLS 2.1  
 Enx-1 BA-RA1C35 NM_007971 Polycomb family, regulate Hox gene expression  2.5 
Kinase       
 PI3 kinase BA-RA5A82 BG354681 Phosphoinositide 3-kinase  
 PKA-like BA-RA4C80 BG354682 cAMP-dependent protein kinase 3.1  
Phosphatase       
 Tyr phosphatase BA-RA1D25 X63440 p19 protein-tyrosine-phosphatase  2.3 B  
RNA binding       
 RBM3 MSC-RA1A25 NM_016809 RNA binding motif 3 4.1  
 Napor-3 BA-RA6B66 AF090697 Apoptosis-related RNA-binding protein 2.7  B  
 Poly(a) binding BA-RA6B35 X65553 RNA-binding protein 2.3  
Protease       
 Calpain 4 MSC-RA17B72 NM_009795 Calcium-dependent protease 6.2  A  
Cell growth       
 Activin beta C MSC-RA15B58 X82540 TGF-beta family  4.3 A  
 TAX responsive MSC-RA15B86 X81987 Malignancy-related protein 107  3.5 A  
 Inhibitor of CDC42 MSC-RA16A75 L07918 Hematopoietic-specific, GDP dissociation inhibitor  21 A  
Protein modification       
 CCT eta MSC-RA12A61 Z31399 Chaperonin containing TCP-1 4.7  
 NEDD-4 MSC-RA16D13 P46935 Containing WW domain 2.1  A  
Membrane associated       
 pRGR1 BA-RA6C77 AF041429 4 TM; unknown function  3.9 
 TM-protein BA-RA4C93 AF082835 Type III transmembrane protein  3.7 B  
 Ly-6C.2 MSC-RA11C15 M18466 u-PAR domain proteins 6.1  A  
Nuclear protein       
 PWP1-like BA-RA5C56 L07758 WD-40, nuclear phosphoprotein  2.3 
 Npm1 BA-RA4A70 NM_008722 Nucleophosmin 1–related protein  
 Karyopherin BA-RA1C82 NM_010655 Importin  5.9 
Calcium binding       
 PW29 BA-RA4C42 D49429 Calcium-binding protein  3.9 B  
 Cam I BA-RA6C79 M19381 Calmodulin-dependent kinase and phosphatase  5.9 B  
Chromosome modeling       
 HAT1 BA-RA6D08 NM_003642 Histone acetyltransferase 1  A/B  
 Histone H2A BA-RA5A12 AF171080 Chromatin structure 3.1  A/B 
 Set beta isoform MSC-RA15B10 S68987 Inhibitor of HAT and PP2a  3.7 
 HMG1-related MSC-RA17A09 U00431 Chromatin organization  A/B 
Miscellaneous       
 Ant2 BA-RA2D75 AF240003 Adenine nucleotide translocase 2  4.1 
 Prg2 MSC-RA14C84 NM_008920 Major basic protein in eosinophils of bone marrow  4.9 A  
 Calponin 2 MSC-RA16D77 NM_007725 Actin binding protein  6.5 
 Transport-like proteins BA-RA1D09 AY008297 RPGR-interacting protein  A/B 
 CYLN2 BA-RA4A42 AAF99333 Cytoplasmic linker proteins  
Novel       
 KIAA0780 BA-RA1D26 BG354685 Unknown function  2.4 B  
 RP42 BA-RA2C82 BG354683 Unknown function  B  
 PTD011 BA-RA5D01 BG354684 Unknown function 2.9  
 Unknown BA-RA3D39 BG354687 Unknown function  2.1 
 Unknown MSC-RA16B18 BG354690 Unknown function 5.1  
 Unknown BA-RA4A52 BG354679 Unknown function  2.3 
 Unknown BA-RA4B58 BG354678 Unknown function  
 Unknown MSC-RA15C76 BG354689 Unknown function  

Libraries A and B were made from Lin−/loSca-1+ and Lin−/loSca-1+Thy-1.1loc-kit+cells, respectively. The changes in expression levels (fold) between Rhlo and Rhhi cells are indicated in the corresponding column of Rhlo and Rhhi. For abbreviations see Table 1.

Table 3.

A partial list of genes commonly expressed in both RhloLin−/loSca-1+c-kit+and RhhiLin−/loSca-1+c-kit+cells

NameClone codeAccession numberDescription
Transcription factor    
 GCN5 BA_RA5B01 AF254441 Mouse histone acetyltransferases 
 Vezf1 BA_RA1B23 NM_016686 Vascular endothelial zinc finger, high homology with DB1 
 Ctcf BA_RA1D45 NM_007794 CCCTC-binding factor, zinc-finger protein 
 Pit-1 BA_RA2A02 S77417 Pituitary-specific POU-domain transcription factor  
 Zfp148 BA_RA2B29 NM_011749 Zinc finger protein 148  
 LIM only 2 (Lmo2) BA_RA2B65 NM_008505 Hemotopoietic-related transcription factors  
 MEF2C BA_RA2C38 NM_002397 MADS/MEF2-family transcription factor  
 BRX BA_RA1A21 Y11896 Breast cancer nuclear receptor-binding auxiliary protein 
 Scmh1 BA_RA2A80 AB03090 Sex comb on midleg homologue protein  
 Fiz1 BA_RA1D58 NM_011813 Flt3 interacting zinc finger protein 1  
 Ciz1 BA_RA3D83 NM_012127 p21 (Cip1/Waf1)-interacting zinc finger protein 
 p53 MSC_RA14D53 AF161020 Cell cycle regulator 
 TOK-1 BA_RA1D59 XM_005841 Cdk inhibitor; p21 binding protein 
Kinase    
 p38 BA_RA5D64 NM_011951 MAP kinase 
Phosphatase    
 P19 BA_RA1D25 NM_009878 Nonreceptor protein tyrosine phosphatase (PTP)  
Nucleic acid binding protein    
 Translin BA_RA5C46 X81464 RNA, DNA-binding protein  
 Cnbp BA_RA6C69 NM_013493 Cellular nucleic acid binding protein 
 Srp20 BA_RA1C83 X91656 RNA-binding protein 
 Nabp MSC_RA12A01 L12693 Nucleic acid binding protein 
Cell growth    
 P40 BA_RA6C60 NM_011029 A subunit of IL-12  
 Cyclophilin MSC_RA13B90 X52803 Secreted growth factor, cyclosporin A–binding protein 
 Stathmin BA_RA1C77 J04797 Also called p19, cytosolic phosphoprotein, developmentally regulated 
 Ptma MSC_RA14D10 NM_008972 Prothymosin alpha, proliferation-related protein  
Protein modification    
 Cct8 BA_RA6B12 NM_009840 Chaperonin subunit 8 (theta)  
 Skp1 BA_RA2C21 AF083214 Skp1/Cull 1/F-box protein (SCF) complex protein Skp1 
 RC7-I MSC_RA12B73 D21799 Beta-type subgroup of proteasomes 
Adhesion    
 Vinexin BA_RA5C01 AF064806 Adhesion associated protein, a vinculin-binding protein with multiple SH3 domains  
 E25B BA_RA5C53 U76253 Integral membrane protein 2B  
Membrane associated    
 Ramp 2 BA_RA2B94 NM_019444 Receptor-activity modifying protein 2 
 Ramp 1 BA_RA1C19 NM_016894 Receptor-activity modifying protein 1  
 TSA-1 MSC_RA12B43 U47737 Thymic shared antigen-1  
 Vamp5 BA_RA2C71 NM_016872 Vesicle-associated membrane protein 5  
Nuclear protein    
 BRCA2 BA_RA6C10 U89652 Breast cancer susceptibility genes  
 RING finger protein MSC_RA15D38 AF022081 Function as a bridging factor and regulate steroid receptor-dependent transcription 
 PTAC97 BA_RA3B79 D45836 Nuclear pore–targeting complex component of 97kDa  
Apoptotic-related protein    
 Survivin BA_RA5C63 AF115517 Inhibitor of apoptosis  
 PCAR BA_RA6B65 AF174394 Apoptotic-related protein  
 Naip7 BA_RA6C21 AF242433 Neuronal apoptosis inhibitory protein  
Histone related    
 Nap1l4 BA_RA6C49 NM_008672 A histone chaperonin  
 H3f3b BA_RA6C53 NM_008211 H3b histone 
Centrosome associated    
 IB3/5 BA_RA5B13 X79131 Polypeptide attached to centrosomes 
Miscellaneous    
 MNCb-3848 BA_RA6B62 AB041599 Brain cDNA, clone MNCb-3848  
 Gdi3 BA_RA1A63 NM_008112 GDP dissociation inhibitor 3 (Gdi3) 
 Septin6 BA_RA1D47 AB023622 A novel member of the structural GTPase subfamily associated with cytokinesis, KIAA0128 is the human counterpart of mouse Septin6; 1 coil 
 Mtprd BA_RA2A46 AB008516 Developmentally regulated gene, involved in Down syndrome  
 Zinc finger protein BA_RA2D28 D10627 Zinc finger protein 
 Unr BA_RA4B25 U40223 Rat unr mRNA for unr protein with unknown function  
 SNX5 MSC_RA11A92 NM_014426 Sorting nexin 5 (SNX5)  
 Transgelin MSC_RA13A39 AF149291 1 calponin homology domain; in thymus 
 ATPase MSC_RA15D42 NM_003945 Lysosomal proton pump 
 BAF57 MSC_RA18A78 AF035263 Mammalian SWI/SNF subunits, critical for chromatin remodeling 
Unknown    
 RP11 BA_RA5D56 AC022766 Unknown function  
 4F5rel BA_RA3B25 AF073297 A candidate phenotypic modifier for SMA 
 EST MSC_RA16B50 BG354688 Unknown function 
 Interferon induced BA_RA5D20 X61381 R. rattus interferon–induced mRNA 
NameClone codeAccession numberDescription
Transcription factor    
 GCN5 BA_RA5B01 AF254441 Mouse histone acetyltransferases 
 Vezf1 BA_RA1B23 NM_016686 Vascular endothelial zinc finger, high homology with DB1 
 Ctcf BA_RA1D45 NM_007794 CCCTC-binding factor, zinc-finger protein 
 Pit-1 BA_RA2A02 S77417 Pituitary-specific POU-domain transcription factor  
 Zfp148 BA_RA2B29 NM_011749 Zinc finger protein 148  
 LIM only 2 (Lmo2) BA_RA2B65 NM_008505 Hemotopoietic-related transcription factors  
 MEF2C BA_RA2C38 NM_002397 MADS/MEF2-family transcription factor  
 BRX BA_RA1A21 Y11896 Breast cancer nuclear receptor-binding auxiliary protein 
 Scmh1 BA_RA2A80 AB03090 Sex comb on midleg homologue protein  
 Fiz1 BA_RA1D58 NM_011813 Flt3 interacting zinc finger protein 1  
 Ciz1 BA_RA3D83 NM_012127 p21 (Cip1/Waf1)-interacting zinc finger protein 
 p53 MSC_RA14D53 AF161020 Cell cycle regulator 
 TOK-1 BA_RA1D59 XM_005841 Cdk inhibitor; p21 binding protein 
Kinase    
 p38 BA_RA5D64 NM_011951 MAP kinase 
Phosphatase    
 P19 BA_RA1D25 NM_009878 Nonreceptor protein tyrosine phosphatase (PTP)  
Nucleic acid binding protein    
 Translin BA_RA5C46 X81464 RNA, DNA-binding protein  
 Cnbp BA_RA6C69 NM_013493 Cellular nucleic acid binding protein 
 Srp20 BA_RA1C83 X91656 RNA-binding protein 
 Nabp MSC_RA12A01 L12693 Nucleic acid binding protein 
Cell growth    
 P40 BA_RA6C60 NM_011029 A subunit of IL-12  
 Cyclophilin MSC_RA13B90 X52803 Secreted growth factor, cyclosporin A–binding protein 
 Stathmin BA_RA1C77 J04797 Also called p19, cytosolic phosphoprotein, developmentally regulated 
 Ptma MSC_RA14D10 NM_008972 Prothymosin alpha, proliferation-related protein  
Protein modification    
 Cct8 BA_RA6B12 NM_009840 Chaperonin subunit 8 (theta)  
 Skp1 BA_RA2C21 AF083214 Skp1/Cull 1/F-box protein (SCF) complex protein Skp1 
 RC7-I MSC_RA12B73 D21799 Beta-type subgroup of proteasomes 
Adhesion    
 Vinexin BA_RA5C01 AF064806 Adhesion associated protein, a vinculin-binding protein with multiple SH3 domains  
 E25B BA_RA5C53 U76253 Integral membrane protein 2B  
Membrane associated    
 Ramp 2 BA_RA2B94 NM_019444 Receptor-activity modifying protein 2 
 Ramp 1 BA_RA1C19 NM_016894 Receptor-activity modifying protein 1  
 TSA-1 MSC_RA12B43 U47737 Thymic shared antigen-1  
 Vamp5 BA_RA2C71 NM_016872 Vesicle-associated membrane protein 5  
Nuclear protein    
 BRCA2 BA_RA6C10 U89652 Breast cancer susceptibility genes  
 RING finger protein MSC_RA15D38 AF022081 Function as a bridging factor and regulate steroid receptor-dependent transcription 
 PTAC97 BA_RA3B79 D45836 Nuclear pore–targeting complex component of 97kDa  
Apoptotic-related protein    
 Survivin BA_RA5C63 AF115517 Inhibitor of apoptosis  
 PCAR BA_RA6B65 AF174394 Apoptotic-related protein  
 Naip7 BA_RA6C21 AF242433 Neuronal apoptosis inhibitory protein  
Histone related    
 Nap1l4 BA_RA6C49 NM_008672 A histone chaperonin  
 H3f3b BA_RA6C53 NM_008211 H3b histone 
Centrosome associated    
 IB3/5 BA_RA5B13 X79131 Polypeptide attached to centrosomes 
Miscellaneous    
 MNCb-3848 BA_RA6B62 AB041599 Brain cDNA, clone MNCb-3848  
 Gdi3 BA_RA1A63 NM_008112 GDP dissociation inhibitor 3 (Gdi3) 
 Septin6 BA_RA1D47 AB023622 A novel member of the structural GTPase subfamily associated with cytokinesis, KIAA0128 is the human counterpart of mouse Septin6; 1 coil 
 Mtprd BA_RA2A46 AB008516 Developmentally regulated gene, involved in Down syndrome  
 Zinc finger protein BA_RA2D28 D10627 Zinc finger protein 
 Unr BA_RA4B25 U40223 Rat unr mRNA for unr protein with unknown function  
 SNX5 MSC_RA11A92 NM_014426 Sorting nexin 5 (SNX5)  
 Transgelin MSC_RA13A39 AF149291 1 calponin homology domain; in thymus 
 ATPase MSC_RA15D42 NM_003945 Lysosomal proton pump 
 BAF57 MSC_RA18A78 AF035263 Mammalian SWI/SNF subunits, critical for chromatin remodeling 
Unknown    
 RP11 BA_RA5D56 AC022766 Unknown function  
 4F5rel BA_RA3B25 AF073297 A candidate phenotypic modifier for SMA 
 EST MSC_RA16B50 BG354688 Unknown function 
 Interferon induced BA_RA5D20 X61381 R. rattus interferon–induced mRNA 

Differential gene expression between HSCs and MPP cells

Since Rh staining allows for separation of HSCs into populations enriched for ST-HSC/early progenitor cells and LT-HSCs,9-11 the Lin−/loSca-1+c-kit+ cells were further separated into Rhlo and Rhhipopulations using flow cytometry (Figure2A-B). In order to avoid a potential interference from the intermediate staining of Rh (Rhitm) cells, we chose a symmetrical portion of cells containing either the 15% highest staining or 15% lowest staining for Rh. A competitive repopulation assay was performed to confirm the functional differences between these 2 populations of cells. As Figure 2C shows, the engraftment rate using Rhlo cells is much higher than that using Rhhi cells after transplantation. In addition, the Rhlo cells could support hematopoiesis for up to 6 months after transplantation (Figure 2C) and were able to reconstitute the bone marrow in the secondary transplantation (F. L. and L. L., data not shown). The Rhhi cells, which gave rise to both myeloid and lymphoid lineages, only engrafted the bone marrow for less than 4 weeks (Figure 2C). This result demonstrated that we had obtained 2 distinct cell populations: the Rhlo population of cells were enriched for HSCs and the Rhhi population of cells were enriched for MPP cells.5 

Fig. 2.

Isolation and functional characterization of Rhlo and Rhhi cell populations.

(A) A cartoon showing 2 important populations of cells: LT-HSCs and multipotent progenitors (MPPs). The LT-HSCs can support long-term (up to 6 months) hematopoiesis in competitive repopulation assay. MPP cells can only briefly support (< 4 weeks) hematopoiesis in the same assay. Rhodamine-123 (Rh) was used to separate these 2 populations of cells. (B) Separation of RhloLin−/loSca-1+c-kit+and RhhiLin−/loSca-1+c-kit+cells using flow cytometry. R3: Lin−/loSca-1+c-kit+ cells; R2: Rhlo (15% of cells with the lowest staining); R4: Rhhi (15% of cells with the highest staining). (C) Competitive repopulation assay. Either RhloLin-/loSca-1+c-kit+or RhhiLin-/loSca-1+c-kit+cells (250 cells) derived from the donor Ly5.2 mice were mixed with 4 × 104 Lin−/lo cells from congenic host strain Ly5.1,17 and injected to lethally irradiated (11 Gy [1100 rads]) Ly5.1 mice. Eight mice were used per group. Peripheral blood cells were harvested at different time points after transplantation as indicated. Flow cytometric analyses were performed using different specific cell surface markers (CD3, B220, Mac-1, and Gr-1) together with anti-Ly5.2 antibody. ○ indicates Rhlo lymphoid lineage; ▵, Rhlo myeloid lineage; ●, Rhhi lymphoid lineage; ▴, Rhhi myeloid lineage.

Fig. 2.

Isolation and functional characterization of Rhlo and Rhhi cell populations.

(A) A cartoon showing 2 important populations of cells: LT-HSCs and multipotent progenitors (MPPs). The LT-HSCs can support long-term (up to 6 months) hematopoiesis in competitive repopulation assay. MPP cells can only briefly support (< 4 weeks) hematopoiesis in the same assay. Rhodamine-123 (Rh) was used to separate these 2 populations of cells. (B) Separation of RhloLin−/loSca-1+c-kit+and RhhiLin−/loSca-1+c-kit+cells using flow cytometry. R3: Lin−/loSca-1+c-kit+ cells; R2: Rhlo (15% of cells with the lowest staining); R4: Rhhi (15% of cells with the highest staining). (C) Competitive repopulation assay. Either RhloLin-/loSca-1+c-kit+or RhhiLin-/loSca-1+c-kit+cells (250 cells) derived from the donor Ly5.2 mice were mixed with 4 × 104 Lin−/lo cells from congenic host strain Ly5.1,17 and injected to lethally irradiated (11 Gy [1100 rads]) Ly5.1 mice. Eight mice were used per group. Peripheral blood cells were harvested at different time points after transplantation as indicated. Flow cytometric analyses were performed using different specific cell surface markers (CD3, B220, Mac-1, and Gr-1) together with anti-Ly5.2 antibody. ○ indicates Rhlo lymphoid lineage; ▵, Rhlo myeloid lineage; ●, Rhhi lymphoid lineage; ▴, Rhhi myeloid lineage.

Close modal

To study differential gene expression among Rhlo and Rhhi cells, we used the microarray glass slides on which the 5000 negatively selected cDNA clones from both libraries A and B had been arrayed in duplicate. These microarrays were hybridized to Cy3- and Cy5-labeled probes generated from Rhlo and Rhhi cells, respectively. Figure3A shows a typical hybridization result showing differentially expressed genes between the Rhlo and the Rhhi cells. Rhlo-Cy3 (green) represents genes that are expressed higher in the enriched HSCs. Rhhi-Cy5 (red) represents genes that are expressed higher in the enriched MPP cells. Yellow represents genes that are expressed in a relatively similar level in both cell populations. Figure 3B shows the results of statistical analysis based on 2 experiments with 4 points (2 points on each slide) representing each cDNA clone, and selection using SpotUnite and SpotSelection software (H. Hammersmark and R.B., see “Materials and methods”). The vertical axis represents the Cy3/Cy5 signal intensity ratio and the horizontal axis was sorted by mean ± standard deviation (sd) (mean indicates average signal intensity of 4 spots for each cDNA insert). We chose cutoff lines that were either 2-fold higher or 2-fold lower for the Cy3/Cy5 intensity ratio. Thus, genes highly expressed in the Rhlo population fell into the upper (positive) region and genes highly expressed in the Rhhi population fell into the lower (negative) region. The spots that fell very close to the left axis represent cDNAs with very weak hybridization signals and, along with spots of significant variation, were ignored. Using this strategy we have identified approximately 30 genes that were highly expressed in the LT-HSCs and around 30 genes that were highly expressed in the MPP cells. These clones were marked with error bars (yellow, Figure 3B). Of these, some were not found in the nonredundant database and therefore were defined as novel or functionally unknown genes (Table 2). To confirm the differential expression of selected genes using microarray analysis, RT-PCR was performed and 15% of the clones were confirmed to be expressed preferentially in HSCs. Activin βC was the only clone from the Rhhi cell group tested that showed predominant expression in MPP cells (Figure 3C).

Fig. 3.

Microarray analysis of HSC gene expression.

(A) Microarray hybridization and spotfinding. Microarray slides described in Figure 1 were hybridized with cDNA probes prepared from cells. Each cDNA probe was labeled with Cy3 and Cy5 fluorophores separately. A combination of these cDNA probes was hybridized with microarrays A (3000 × 2) and B (2000 × 2). A typical hybridization result was shown using the Cy3-labeled cDNA probe from RhloLin−/loSca-1+c-kit+and the Cy5-labeled probe from RhhiLin−/loSca-1+c-kit+cells. (B) Statistical analysis and selection. A typical example showing the microarray hybridization results obtained from 2 experiments scanned by Genescanner (Molecular Dynamics) and analyzed by SpotUnite and SpotSelection programs (see text for details). (C) RT-PCR assays. To confirm the differential expression of genes between the Rhlo and Rhhi cells, 15% candidate genes were analyzed by RT-PCR as described in “Materials and methods.”

Fig. 3.

Microarray analysis of HSC gene expression.

(A) Microarray hybridization and spotfinding. Microarray slides described in Figure 1 were hybridized with cDNA probes prepared from cells. Each cDNA probe was labeled with Cy3 and Cy5 fluorophores separately. A combination of these cDNA probes was hybridized with microarrays A (3000 × 2) and B (2000 × 2). A typical hybridization result was shown using the Cy3-labeled cDNA probe from RhloLin−/loSca-1+c-kit+and the Cy5-labeled probe from RhhiLin−/loSca-1+c-kit+cells. (B) Statistical analysis and selection. A typical example showing the microarray hybridization results obtained from 2 experiments scanned by Genescanner (Molecular Dynamics) and analyzed by SpotUnite and SpotSelection programs (see text for details). (C) RT-PCR assays. To confirm the differential expression of genes between the Rhlo and Rhhi cells, 15% candidate genes were analyzed by RT-PCR as described in “Materials and methods.”

Close modal

The differentially expressed genes fall into a variety of gene families (Table 2). Among the genes that are highly expressed in HSCs are several clones identified as transcription factors. Nfix belongs to the conserved nuclear factor I (NFI) family of transcription/replication proteins. Loss of function of Nfia, one of 4 Nfix family members, causes severe developmental defects, suggesting this family may have distinct roles in vertebrate development.31 Two novel genes that encode a PI3 kinase and a PKA kinase were also identified. The homologue of PKA-like kinase in Schizosaccharomyces pombe, Kin, was found to be important for growth polarity.32 PI3 kinase can be activated by c-kit/stem cell factor receptor signaling and is essential for male fertility.33 Furthermore, PI3 kinase has been known to be essential for cell survival.34 RBM3, an RNA-binding protein, is up-regulated by granulocyte macrophage–colony-stimulating factor (GM-CSF)35 and is closely related to the Y chromosome ribonucleic acid recognition motif (YRRM).36YRRM has been implicated in azoospermia, a male infertility disease, and spermatogenesis.36 This suggests that RBM3 might be involved in the regulation of HSC development. Histone acetyltransferase 1 (HAT1) is also highly expressed in the Rhlo cells. HAT1 belongs to the GCN5-related N-acetyltansferase superfamily and functions to acetylate histone H4, thereby participating in chromatin-related transcriptional regulation.37 Recently HAT1 was found to be involved with telomeric silencing by the transcriptional repression of telomere-proximal genes.38 In contrast, Set oncoprotein, a component of a recently identified protein complex, INHAT, that inhibits the activity of HAT,39 was highly expressed in the Rhhi cell population.

Among the genes that are highly expressed in Rhhi cells is the growth factor Activin βC. Activin βC belongs to the transforming growth factor β (TGF-β) family, which functions in the determination of cell fate and pattern formation during embryogenesis.40 However, the function of Activin βC remains unknown. Other members of the Activin family (A and B) are known to function as dimeric proteins with diverse biologic activities in vertebrate reproduction.41 Expression of Activin βC in the MPP cells suggests that it may be involved in the regulation of HSC differentiation and proliferation. Another gene differentially expressed in the MPP population is Enx-1, a mouse homologue of Drosophila enhancer of zeste and member of the Polycomb family of transcriptional regulators.42 It controls the expression of several homeobox genes. The HoxA9gene was also identified in the Rhhi cells. HoxA9 was found to be involved in translocation t(7;11)(p15;p15), present in acute myeloid leukemia (AML) French-American-British (FAB) classification M2-M4.43 Inactivation ofHoxA9 was shown to have defects in myeloid, erythroid, and lymphoid development in knockout mice.44 Overexpression ofHoxA9 and Meis1 has been shown to induce murine leukemia45 and overexpression of HoxA9 alone can immortalize myeloid progenitors.46 

We also identified approximately 70 genes that are expressed in both LT-HSCs and MPP cells (Table 3). Among these is the transcription factor GCN5, a histone acetyltransferase, which interacts with Notch 1 and plays an important role in the recombination recognition sequence binding protein J (RBP-J)–mediated transactivation by Notch 1.47 Loss of GCN5 leads to increased apoptosis and mesodermal defects during mouse development.48 Fiz1, a zinc finger protein with 11 C2H2-type zinc fingers, interacts with receptor tyrosine kinase Flt3.49 Flt3 has been shown to play a role in proliferation and survival of hematopoietic progenitor cells as well as differentiation of early B-lymphoid progenitors, dendritic cells, and natural killer cells.50 TOK-1, a POU-domain–containing transcription factor, is a p21 C-terminal–binding protein and preferentially binds to an active form of cyclin-dependent kinase 2 (CDK2) via p21.51 p21 is a negative cell-cycle regulator and is involved in maintaining the quiescent state of HSCs.52 One of the identified apoptosis-related molecules is survivin. Survivin is an inhibitor of apoptosis and is overexpressed in various human cancers but undetectable in normal tissues.53 Overexpression of survivin resulted in an accelerated S phase shift, resistance to G1 arrest, and activated Cdk2/Cyclin E complex leading to Rb phosphorylation.54 

The mechanisms that regulate the self-renewal of HSCs are largely unknown. The deterministic model of hematopoiesis suggests that HSC self-renewal is autonomous; thus, expression of specific sets of genes could determine the stem cell fate. Therefore, a systematic approach was used to study differential gene expression profiles in adult HSCs. The ability to isolate a cell population enriched with HSCs allowed us to study the gene expression profile of HSCs. Using macroarray and microarray techniques, we were able to obtain a snapshot of genes expressed by enriched populations of HSCs as well as MPP cells. The identification of multiple genes that regulate proliferation, cell survival, immortalization, and differentiation gives new insights into HSC functions. This is important not only to study their biologic behaviors but also for potential clinical applications. For example, the efficacy of treatment modalities for cancer, such as radiation therapy and many chemotherapy agents, is constrained by dose-limiting bone marrow toxicity. Therefore, the ability to isolate HSCs with self-renewal and repopulating potentials, in vitro amplification of these cells, and transplantation without tumor cells would greatly increase the success rate of current cancer therapies.

Ex vivo–expanded stem cells have been extensively tested for possible use in transplantation. Soluble jagged-1, a Notch ligand, induced the survival and expansion of human stem cells with multipotent repopulating capacity.55 The presence of Notch 1 in the HSC library suggests that these proteins may play a role in self-renewal or maintaining identity of HSCs. Although various growth factors can induce stem cells to proliferate in vitro, they result in concomitant differentiation. The function of stem cell factor (SCF) has been known as a hematopoietic stem cell survival factor.56Recently, it has also been shown that the in vitro culture of the HSCs, in the presence of both SCF and thrombopoietin (TPO), induced self-renewal cell division in which only one of the daughter cells had self-renewal potential.57 Asymmetric cell division requires unequal segregation of cell-fate determinants, such as mRNAs and proteins, which are important to maintain self-renewal and repopulating potentials, during mitosis. This type of asymmetric cell division is also observed in germline cell division. DuringDrosophila oogenesis, a germline stem cell divides asymmetrically to produce a daughter stem cell and a cytoblast, which further divides to eventually produce oocytes. This process is controlled by several factors including decapentaplegic (DPP)58 as well as by intrinsic mechanisms involving pumilio, nanos, arrest, and bag-of-marbles. Expression of BMP4, the mammalian homologue of DPP, in HSCs (Table 1) suggests that BMP4 may also play a similar role.

The differential gene expression pattern between Rhloand Rhhi indicated that some of the genes were either HSC- or MPP-specific (Table 2), and many of them were expressed in both populations of cells (Table 3). Some of the MPP highly expressed genes included Activin βC, nuclear molecules (PWP-1, Npm1, and karyopherin), TAX-responsive factor, and inhibitor of CDC42. CDC42 was reported to play a role in controlling HSC shape, adhesion, migration, and mobilization.59 Genes that are preferentially expressed in HSCs include transcription factors, RNA-binding proteins, chromatin modifiers, and protein kinases, many of which are involved in developmental regulation. The HSC preferentially expressed genes might be candidate genes that play a role in stem cell self-renewal.

In summary, cDNA libraries were generated from mouse HSCs with long-term reconstituting potential and from progenitor cells, and the differential gene expression patterns were studied using bioinformatics and array technologies. Although the microarray system has been known for its tendency to lose less abundant mRNAs due to limited resolution, our approach overcomes this tendency by removing cDNAs that show high hybridization signals using macroarray and thereby enriching for cDNAs derived from less-abundant mRNAs. On the other hand, our subtraction technique using cDNA probes derived from mature populations of cells may lead to a loss of molecules that are expressed in multiple developmental stages. The systematic approach used in this study provides an effective and efficient method for identification of genes that are specifically expressed in HSCs with self-renewal capability. This study will therefore provide a fundamental tool for identifying important candidate genes involved in the regulation of self-renewal and expansion of HSCs.

We thank G. van den Engh, K. Allen, and D. Corden for the assistance of cell sorting. We thank E. Hammersmark for the assistance on microarray analysis software. We appreciate V. Ng and X. Tao for their assistance on analysis of Est sequences. We thank A. Banta for the involvement of macroarray work. We are grateful to Drs G. Vassilopoulous, D. Russel, and N. Wolf for the consultation on the bone marrow transplantation and repopulation assay. We appreciate the help of R. Krumlauf, B. Steenhard, and D. Stenger on the manuscript proofreading. We are particularly grateful to X. He, J. Zhang, and C. Niu for their work on the reanalysis of cDNA sequences listed in the tables.

Supported in part by National Institutes of Health grant 1P01 DK53074-02 and Stowers Institute for Medical Research.

I.-K.P. and Y.H. contributed equally to this article.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Morrison
SJ
Uchida
N
Weissman
IL
The biology of hematopoietic stem cells.
Annu Rev Cell Dev Biol.
11
1995
35
71
2
Uchida
N
Weissman
IL
Searching for hematopoietic stem cells: evidence that Thy-1.1lo Lin- Sca-1+ cells are the only stem cells in C57BL/Ka-Thy-1.1 bone marrow.
J Exp Med.
175
1992
175
184
3
Spangrude
GJ
Heimfeld
S
Weissman
IL
Purification and characterization of mouse hematopoietic stem cells.
Science.
241
1988
58
62
4
Lagasse
E
Shizuru
JA
Uchida
N
Tsukamoto
A
Weissman
IL
Toward regenerative medicine.
Cell.
14
2001
425
436
5
Morrison
SJ
Weissman
IL
The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype.
Immunity.
1
1994
661
673
6
Kondo
M
Weissman
IL
Akashi
K
Identification of clonogenic common lymphoid progenitors in mouse bone marrow.
Cell.
91
1997
661
672
7
Pohlmann
SJ
Slayton
WB
Spangrude
GJ
Stem cell populations: purification and behavior.
Hematopoiesis—A Developmental Approach. Oxford
Zon
LI
2001
35
47
Oxford University Press
United Kingdom
8
Weissman
IL
Translating stem and progenitor cell biology to the clinic: barriers and opportunities.
Science.
287
2000
1442
1446
9
Spangrude
GJ
Johnson
GR
Resting and activated subsets of mouse multipotent hematopoietic stem cells.
Proc Natl Acad Sci U S A.
87
1990
7433
7437
10
Wolf
NS
Kone
A
Priestley
GV
Bartelmez
SH
In vivo and in vitro characterization of long-term repopulating primitive hematopoietic cells isolated by sequential Hoechst 33342-rhodamine 123 FACS selection.
Exp Hematol.
21
1993
614
622
11
Zijlmans
JM
Visse
JW
Kleiverda
K
Kluin
PM
Willemze
R
Fibbe
WE
Modification of rhodamine staining allows identification of hematopoietic stem cells with preferential short-term or long-term bone marrow-repopulating ability.
Proc Natl Acad Sci U S A.
92
1995
8901
8905
12
Chaudhary
PM
Roninson
IB
Expression and activity of P-glycoprotein, a multidrug efflux pump, in human hematopoietic stem cells.
Cell.
66
1991
85
94
13
Cheshier
SH
Morrison
SJ
Liao
X
Weissman
IL
In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells.
Proc Natl Acad Sci U S A.
96
1999
3120
3125
14
Dexter
TM
Moore
MA
Sheridan
AP
Maintenance of hemopoietic stem cells and production of differentiated progeny in allogeneic and semiallogeneic bone marrow chimeras in vitro.
J Exp Med.
145
1977
1612
1616
15
Park
IK
Klug
CA
Li
K
et al
Molecular cloning and characterization of a novel regulator of G-protein signaling from mouse hematopoietic stem cells.
J Biol Chem.
276
2001
915
923
16
Geiss
GK
Bumgarner
RE
An
MC
et al
Large-scale monitoring of host cell gene expression during HIV-1 infection using cDNA microarrays.
Virology.
266
2000
8
16
17
Osawa
M
Hanada
K
Hamada
H
Nakauchi
H
Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell.
Science.
273
1996
242
245
18
Maier
E
Meier-Ewert
S
Ahmadi
AR
Curtis
J
Lehrach
H
Application of robotic technology to automated sequence fingerprint analysis by oligonucleotide hybridisation.
J Biotechnol.
35
1994
191
203
19
Schena
M
Shalon
D
Davis
RW
Brown
PO
Quantitative monitoring of gene expression patterns with a complementary DNA microarray.
Science.
270
1995
467
470
20
Spangrude
GJ
Brooks
DM
Tumas
DB
Long-term repopulation of irradiated mice with limiting numbers of purified hematopoietic stem cells: in vivo expansion of stem cell phenotype but not function.
Blood.
85
1995
1006
1016
21
Jacobs
JJ
Kieboom
K
Marino
S
DePinho
RA
van Lohuizen
M
The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus.
Nature.
397
1999
164
168
22
Kiyono
T
Foster
SA
Koop
JI
McDougall
JK
Galloway
DA
Klingelhutz
AJ
Both Rb/p16INK4a inactivation and telomerase activity are required to immortalize human epithelial cells.
Nature.
396
1998
84
88
23
Morrison
SJ
Prowse
KR
Ho
P
Weissman
IL
Telomerase activity in hematopoietic cells is associated with self-renewal potential.
Immunity.
5
1996
207
216
24
Li
L
Milner
L
Deng
Y
et al
Human homolog of rat Jagged 1 expressed by marrow stroma inhibits differentiation of 32D cells through interaction with Notch1.
Immunity.
8
1998
43
55
25
Jones
P
May
G
Healy
L
et al
Stromal expression of Jagged 1 promotes colony formation by fetal hematopoietic progenitor cells.
Blood.
92
1998
1505
1511
26
Varnum-Finney
B
Purton
LE
Yu
M
et al
The Notch ligand, Jagged 1, influences the development of primitive hematopoietic precursor cells.
Blood.
91
1998
4084
4091
27
Varnum-Finney
B
Xu
L
Brashem-Stein
C
et al
Pluripotent, cytokine-dependent, hematopoietic stem cells are immortalized by constitutive notch1 signaling.
Nat Med.
6
2000
1278
1281
28
Lau
YF
Chan
KM
Sparkes
R
Male-enhanced antigen gene is phylogenetically conserved and expressed at late stages of spermatogenesis.
Proc Natl Acad Sci U S A.
86
1989
8462
8466
29
Phillips
RL
Ernst
RE
Brunk
B
et al
The genetic program of hematopoietic stem cells.
Science.
288
2000
1635
1640
30
Geschwind
DH
Ou
J
Easterday
MC
et al
A genetic analysis of neural progenitor differentiation.
Neuron.
29
2001
325
339
31
das Neves
L
Duchala
CS
Godinho
F
et al
Disruption of the murine nuclear factor I-A gene (Nfia) results in perinatal lethality, hydrocephalus, and agenesis of the corpus callosum.
Proc Natl Acad Sci U S A.
96
1999
11946
11951
32
Levin
DE
Bishop
JM
A putative protein kinase gene (kin1+) is important for growth polarity in Schizosaccharomyces pombe.
Proc Natl Acad Sci U S A.
87
1990
8272
8276
33
Jiang
G
Hyman
R
Lee
KF
Gorman
SO
Blume-Jensen
P
Hunter
T
Kit/stem cell factor receptor-induced activation of phosphatidylinositol 3′-kinase is essential for male fertility.
Nature Genet.
24
2000
157
162
34
Blume-Jensen
P
Janknecht
R
Hunter
T
The kit receptor promotes cell survival via activation of PI 3-kinase and subsequent Akt-mediated phosphorylation of Bad on Ser136.
Curr Biol.
8
1998
779
782
35
Baghdoyan
S
Dubreuil
P
Eberle
F
Gomez
S
Capture of cytokine-responsive genes (NACA and RBM3) using a gene trap approach.
Blood.
95
2000
3750
3757
36
Derry
JM
Kerns
JA
Francke
U
RBM3, a novel human gene in Xp11.23 with a putative RNA-binding domain.
Hum Mol Genet.
4
1995
2307
2311
37
Dutnall
RN
Tafrov
ST
Sternglanz
R
Ramakrishnan
V
Structure of the histone acetyltransferase Hat1: a paradigm for the GCN5-related N-acetyltransferase superfamily.
Cell.
94
1998
427
438
38
Kelly
TJ
Qin
S
Gottschling
DE
Parthun
MR
Type B histone acetyltransferase Hat1p participates in telomeric silencing.
Mol Cell Biol.
20
2000
7051
7058
39
Sang-beom
S
McNamara
P
Heo
S
Turner
A
Lane
WS
Chakravarti
D
Regulation of histone acetylation and transcription by INHAT, a human cellular complex containing the Set oncoprotein.
Cell.
104
2001
119
130
40
Padgett
RW
TGFbeta signaling pathways and human diseases.
Cancer Metastasis Rev.
18
1999
247
259
41
Ethier
JF
Findlay
JK
Roles of activin and its signal transduction mechanisms in reproductive tissues.
Reproduction.
121
2001
667
675
42
Hobert
O
Sures
I
Ciossek
T
Fuchs
M
Ullrich
A
Isolation and developmental expression analysis of Enx-1, a novel mouse Polycomb group gene.
Mech Dev.
55
1996
171
184
43
Borrow
J
Stanton
VP
Andresen
JM
et al
The t(7;11)(p15;p15) translocation in acute myeloid leukaemia fuses the genes for nucleoporin NUP98 and class I homeoprotein HOXA9.
Nat Genet.
12
1996
159
167
44
Lawrence
HJ
Helgason
CD
Sauvageau
G
et al
Mice bearing a targeted interruption of the homeobox gene HOXA9 have defects in myeloid, erythroid, and lymphoid hematopoiesis.
Blood.
89
1997
1922
1930
45
Lawrence
HJ
Rozenfeld
S
Cruz
C
et al
Frequent co-expression of the HOXA9 and MEIS1 homeobox genes in human myeloid leukemias.
Leukemia.
13
1999
1993
1999
46
Schnabel
CA
Jacobs
Y
Cleary
ML
HoxA9-mediated immortalization of myeloid progenitors requires functional interactions with TALE cofactors Pbx and Meis.
Oncogene.
19
2000
608
616
47
Kurooka
H
Honjo
T
Functional interaction between the mouse notch1 intracellular region and histone acetyltransferases PCAF and GCN5.
J Biol Chem.
275
2000
17211
17220
48
Xu
W
Edmondson
DG
Evrard
YA
Wakamiya
M
Behringer
RR
Roth
SY
Loss of Gcn5l2 leads to increased apoptosis and mesodermal defects during mouse development.
Nat Genet.
26
2000
229
232
49
Wolf
I
Rohrschneider
LR
Fiz1, a novel zinc finger protein interacting with the receptor tyrosine kinase Flt3.
J Biol Chem.
274
1999
21478
21484
50
McKenna
HJ
Stocking
KL
Miller
RE
et al
Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells.
Blood.
95
2000
3489
3497
51
Ono
T
Kitaura
H
Ugai
H
et al
TOK-1, a novel p21Cip1-binding protein that cooperatively enhances p21-dependent inhibitory activity toward CDK2 kinase.
J Biol Chem.
275
2000
31145
31154
52
Cheng
T
Rodrigues
N
Shen
H
et al
Hematopoietic stem cell quiescence maintained by p21cip1/waf1.
Science.
287
2000
1804
1808
53
Adida
C
Recher
C
Raffoux
E
Daniel
MT
et al
Expression and prognostic significance of survivin in de novo acute myeloid leukaemia.
Br J Haematol.
111
2000
196
203
54
Li
F
Ambrosini
G
Chu
EY
et al
Control of apoptosis and mitotic spindle checkpoint by survivin.
Nature.
396
1998
580
584
55
Karanu
FN
Murdoch
B
Gallacher
L
et al
The notch ligand jagged-1 represents a novel growth factor of human hematopoietic stem cells.
J Exp Med.
192
2000
1365
1372
56
Brandt
JE
Bhalla
K
Hoffman
R
Effects of interleukin-3 and c-kit ligand on the survival of various classes of human hematopoietic progenitor cells.
Blood.
83
1994
1507
1514
57
Ema
H
Takano
H
Sudo
K
Nakauchi
H
In vitro self-renewal division of hematopoietic stem cells.
J Exp Med.
192
2000
1281
1288
58
Xie
T
Spradling
AC
Decapentaplegic is essential for the maintenance and division of germline stem cells in the Drosophila ovary.
Cell.
94
1998
251
260
59
Yang
FC
Atkinson
SJ
Gu
Y
et al
Rac and Cdc42 GTPases control hematopoietic stem cell shape, adhesion, migration, and mobilization.
Proc Natl Acad Sci U S A.
98
2001
5614
5618

Author notes

Linheng Li, Stowers Institute for Medical Research, 1000 E 50th St, Kansas City, MO 64110; e-mail:lil@stowers-institute.org.

Sign in via your Institution