To clarify whether the expression of the WT1 gene in leukemic cells is aberrant or merely reflects that in normal counterparts, the expression levels of the WT1 gene were quantitated for normal hematopoietic progenitor cells. Bone marrow (BM) and umbilical cord blood (CB) cells were fluorescence-activated cell sorting (FACS)-sorted into CD34+ and CD34 cell populations, and the CD34+ cells into nine subsets (CD34+CD33, CD34+CD33+, CD34+CD38, CD34+CD38+, CD34+HLA-DR, CD34+HLA-DR+, CD34+c-kithigh, CD34+c-kitlow, and CD34+c-kit) according to the expression levels of CD34, CD33, CD38, HLA-DR, and c-kit. Moreover, acute myeloid leukemic cells were also FACS-sorted into four populations (CD34+CD33, CD34+CD33+, CD34 CD33+, and CD34 CD33). FACS-sorted normal hematopoietic progenitor and leukemic cells and FACS-unsorted leukemic cells were examined for the WT1 expression by quantitative reverse transcriptase-polymerase chain reaction. The WT1 expression in the CD34+ and CD34 cell populations and in the nine CD34+ subsets of BM and CB was at either very low (1.0 to 2.4 × 10−2) or undetectable (<10−2) levels (the WT1 expression level of K562 cells was defined as 1.0), whereas the average levels of WT1 expression in FACS-sorted and -unsorted leukemic cells were 2.4 to 9.3 × 10−1. Thus, the WT1 expression levels in normal hematopoietic progenitor cells were at least 10 times less than those in leukemic cells. Therefore, we could not find any normal counterparts of BM or CB that expressed the WT1 at levels comparable with those in leukemic cells. These results indicate an aberrant overexpression of the WT1 gene in leukemic cells and imply the involvement of this gene in human leukemogenesis.

UNLIKE TUMOR SUPPRESSOR genes Rb and p53, which are expressed ubiquitously, the expression of the WT1 gene is restricted to a limited set of tissues such as fetal kidney, ovary, testis, spleen, and the mesothelial cell lining of visceral organs.1,2 Analysis of mice carrying disrupted WT1 gene showed a crucial role of the WT1 gene in early urogenital development.3 In the hematopoietic system, WT1 mRNA was detected at high levels in spleen and at low levels (10−4; the WT1 expression level in K562 cells was defined as 1.0) in bone marrow (BM). WT1 expression in spleen was found to be restricted to the splenic capsule and stroma, but was not detected in splenocytes.2 However, little is known about WT1 expression in normal hematopoietic progenitor cells.

Concerning the involvement of the WT1 gene in human malignancies other than Wilms tumor, it has been reported that the WT1 gene was mutated in a human mesothelioma2 and that some human leukemia samples expressed the WT1 gene.4 We recently found that all 96 human leukemia samples examined by us expressed the WT1 gene, regardless of the disease subtype (acute myelogenous leukemia [AML], acute lymphocytic leukemia [ALL], or chronic myelogenous leukemia [CML]) as a result of raising the detection sensitivity of WT1 mRNA by means of reverse transcriptase-polymerase chain reaction (RT-PCR) assay.5 We showed in the same study that the WT1 expression levels increased in parallel with the progression of clinical stage in CML and correlated inversely with the prognosis for acute leukemia.5 Thus, WT1 was identified as a new prognostic factor and a new marker for the diagnosis of minimal residual disease in human leukemia.5,6 Subsequently, we found that WT1 antisense oligomers inhibited the growth of fresh human leukemic cells and leukemic cell lines, but not that of normal colony-forming unit granulocyte-macrophage (CFU-GM), suggesting an important role of WT1 in leukemic cell growth.7 However, it is unknown whether the WT1 expression in leukemic cells is aberrant or merely reflects that in normal counterparts.

In the present study, we examined the expression levels of the WT1 gene in fluorescence-activated cell sorting (FACS)-sorted hematopoietic progenitor cells of BM and cord blood (CB) to determine whether or not the WT1 overexpression in leukemic cells is aberrant. We describe here that the WT1 expression in normal hematopoietic progenitor cells of BM and CB is either very low or undetectable and at least 10 times less than that in leukemic cells. The role of the WT1 gene in human leukemogenesis is also discussed.

Cell lines.SAM-1 and BV173 (derived from CML, blastic crisis) were kindly provided by Dr E. Tatsumi (Kobe University, Hyogo, Japan) and Dr J. Minowada (Hayashibara Biochemical Labs, Inc, Okayama, Japan), respectively. Kasumi-1 (derived from t[8; 21] AML-M2), was kindly provided by Dr K. Kita (Mie University, Mie, Japan), TF-1 (derived from AML-M6) by Dr T. Kitamura (DNAX Research Institute, Palo Alto, CA), DL-40 (derived from non-Hodgkin's lymphoma, diffuse large-cell type) by Dr I. Kubanishi (Kochi Medical School, Kochi, Japan), and KT-3 (interleukin-6 [IL-6]–dependent T-cell lymphoma cell line) by Dr S. Shimizu (Kanazawa Medical University, Ishikawa, Japan). The remaining cell lines were kindly provided by the Japanese Cancer Research Resources Bank (Tokyo, Japan). The cell lines were cultured in RPMI 1640 medium containing 10% heat-inactivated fetal bovine serum (FBS).

Cell preparation.After informed consent was obtained, BM cells were collected by aspiration from the posterior iliac crest of healthy adult volunteers. Umbilical CB was obtained after normal vaginal deliveries at full term by aspiration from pregnant women from whom informed consent was also obtained. BM aspirate was placed in a 16-mL Falcon tissue culture tube and CB in a 50-mL one containing 400 U of preservative-free heparin (Shimizu Pharmaceutical Co, Osaka, Japan). Mononuclear cells were separated by using Ficoll-Paque (Pharmacia, Piscataway, NJ) density gradient centrifugation, and nonadherent cells were recovered by overnight adherence to plastic dishes. The mononuclear nonadherent cell fractions were further enriched by negative selection using soybean agglutinin (SBA) Micro-CELLector Flasks (Applied Immune Science Inc, Menlo Park, CA) according to the manufacturer's instructions. Leukemic cells were isolated by aspiration from BM or peripheral blood (PB) of patients with leukemia, and their mononuclear cell fractions were obtained as described above. For some of the experiments, previously frozen mononuclear cells were used after rapid thawing.

Purification of hematopoietic progenitor and leukemic cells by cell sorting.Highly purified progenitors were separated from BM- or CB- derived SBA cells by using a FACStar Plus (Becton Dickinson [BD] Immunocytometry Systems, San Jose, CA) equipped with an argon laser tuned at 488 nm, as reported elsewhere.8,9 BM- or CB-derived SBA cells were washed twice with phosphate-buffered saline (PBS)(-) containing 2% FBS (staining medium) and were passed through a stainless-steel mesh. The cells were then pelleted before staining with monoclonal antibodies (MoAbs). The following MoAbs were used: fluorescein isothiocyanate (FITC)-conjugated HPCA-2 (mouse IgG1 CD34 MoAb; BD); phycoerythrin (PE)-conjugated HLA-DR (mouse IgG2a; BD); PE-conjugated CD33 (MY9; mouse IgG2b; Coulter Immunology, Hialeah, FL); PE-conjugated CD38 (mouse IgG1; BD); purified antihuman c-kit MoAb (mouse IgM; Immunotech S.A., Marseille Cedex, France); and goat antimouse IgM with PE (Immunotech S.A.). For double staining, cells were incubated with 20 μL of HLA-DR, CD33, or CD38/106 cells for 30 minutes on ice and then washed twice with staining medium. Cells were also stained with 20 μL of purified antihuman c-kit MoAb/5 × 105 cells for 30 minutes at room temperature. After washing with staining medium, cells were incubated with 20 μL of goat antimouse IgM with PE/5 × 105 cells for 30 minutes at room temperature. These cells were then washed twice with staining medium and stained with HPCA-2 (FITC) as described elsewhere.8,9 All the stained cells were kept on ice until cell sorting. For negative controls, unstained cells and cells stained only with second antibody or with isotype control IgG with either FITC or PE were included.

Stained cells were sorted with a FACStar Plus single-laser flow cytometry system. Sorting gates were established for both forward scattering and side scattering, and dual-parameter dotograms showing FITC (CD34) and PE (CD33, CD38, HLA-DR, or c-kit) fluorescence were generated from the gated events. These gated dotograms were then used to sort the cells into CD34+CD33+/−, CD34+CD38+/−, or CD34+HLA-DR+/− cells. The sorting windows for CD34+ cells expressing different levels of c-kit protein were also established as previously reported.9 CD34+c-kithigh, low or − cells were then separately sorted. Typical scattergrams of BM-, CB-, or leukemia-derived CD34+ cells expressing different levels of CD33, CD38, HLA-DR, or c-kit are presented in Figs 1, 2, and 3. Data were aquired with FACStar Plus Research Software, and a minimum of 20,000 events was analyzed for each sample. After sorting, the recovered cells were washed twice with α-medium, then washed with ice-cold PBS(-) and stored in D solution at −80°C for the detection of WT1 mRNA by RT-PCR. The purity of FACS-sorted cells of BM and CB was greater than 90%.

Fig. 1.

Expression of CD33, CD38, HLA-DR, or c-kit on BM- derived CD34+ cells (A) or CB-derived CD34+ cells (B). Sorting gates were first established for forward scattering and side scattering. Four dual-parameter dotograms displaying FITC (CD34) and PE (CD33, CD38, HLA-DR, or c-kit) fluorescence were then generated from the gated events. (C) Leukemic cells obtained from a patient with AML (M4, patient no. 294) were sorted as described. A dual-parameter dotogram displaying CD34 and CD33 is shown.

Fig. 1.

Expression of CD33, CD38, HLA-DR, or c-kit on BM- derived CD34+ cells (A) or CB-derived CD34+ cells (B). Sorting gates were first established for forward scattering and side scattering. Four dual-parameter dotograms displaying FITC (CD34) and PE (CD33, CD38, HLA-DR, or c-kit) fluorescence were then generated from the gated events. (C) Leukemic cells obtained from a patient with AML (M4, patient no. 294) were sorted as described. A dual-parameter dotogram displaying CD34 and CD33 is shown.

Close modal
Fig. 2.

Expression of the WT1 gene in FACS-sorted normal BM (A) or leukemic (B) cells. Representative cases are shown. To detect WT1 mRNA, PCR was performed for 45 cycles (35 and 10 cycles for first and second round PCR, respectively). To quantitate β-actin mRNA, PCR was performed for 31 cycles.

Fig. 2.

Expression of the WT1 gene in FACS-sorted normal BM (A) or leukemic (B) cells. Representative cases are shown. To detect WT1 mRNA, PCR was performed for 45 cycles (35 and 10 cycles for first and second round PCR, respectively). To quantitate β-actin mRNA, PCR was performed for 31 cycles.

Close modal
Fig. 3.

Expression levels of the WT1 gene in normal hematopoietic progenitor and leukemic cells. Expression levels of the WT1 gene in K562 cells were defined as 1.0.

Fig. 3.

Expression levels of the WT1 gene in normal hematopoietic progenitor and leukemic cells. Expression levels of the WT1 gene in K562 cells were defined as 1.0.

Close modal

RT-PCR.Quantitative RT-PCR of FACS-sorted cells was performed as described previously5,10 with some modifications. Briefly, total RNA was extracted from 2,000 to 4,000 cells with the acid guanidine-phenol chloroform method and then converted into cDNA on a scale of 20 μL. The expression level of the WT1 gene in K562 cells was defined as 1.0 as described previously.5 PCR was performed for 35 cycles with the outer primers to quantitate 100 to 10−2 levels of WT1 expression. For the quantitation of 10−2 to 10−3 levels of the WT1 expression, a second PCR using nested internal primers was performed for 10 to 14 cycles. To quantitate β-actin levels, PCR was also performed for 30 to 32 cycles. To normalize the differences in RNA degradation for individual samples and in RNA loading for RT-PCR, the value of the WT1 gene expression divided by the β-actin gene expression was defined as the WT1 expression levels of the samples. Serial 1:10 dilutions of standard cDNAs prepared from 2,000 K562 cells were always PCR-amplified simultaneously with the samples to determine the WT1 expression levels of the samples relative to those of K562 cells. Because the amount of total RNA that could be extracted was very small because of the small number of FACS-sorted cells, the detection limit of WT1 mRNA by RT-PCR was restricted to the 10−2 level. Quantitative RT-PCR of fresh leukemic cells and leukemic cell lines was performed as described previously5,10 with some modifications. Briefly, 2.0 μg of total RNA from leukemic samples was converted into cDNA in 30 μL of reaction buffer. PCR was performed for 27 to 30 cycles for the quantitation of WT1 mRNA and for 18 cycles for the quantitation of β-actin mRNA. All experiments were performed in duplicate.

Very low or undetectable levels of WT1 expression in hematopoietic progenitor cells of BM and umbilical CB.BM and umbilical CB cells were FACS-sorted according to the levels of the expression of CD34, CD33, CD38, HLA-DR, and c-kit (Figs 1A and B). CD34+ and CD34 cell populations, and nine CD34+ subsets of CD34+CD33, CD34+CD33+, CD34+CD38, CD34+CD38+, CD34+HLA-DR, CD34+HLA-DR+, CD34+c-kithigh, CD34+c-kitlow, and CD34+ c-kit were isolated and examined for the expression levels of the WT1 gene (Figs 2 and 3, Table 1). The WT1 expression levels of CD34+ and CD34 cell populations and of the nine CD34+ subsets of BM and CB were all very low (1.0 to 2.4 × 10−2) or undetectable (<10−2). Of 5 CD34+ cell samples (3 and 2 samples from BM and CB, respectively), 4 expressed very low levels (1.0 to 2.4 × 10−2) of WT1, whereas the WT1 expression in all the 5 CD34 cell samples (3 and 2 samples from BM and CB, respectively) were at undetectable levels (<10−2). Thus, the WT1 expression levels were significantly (P < .01) higher in CD34+ cells than in CD34 cells. No significant difference in the WT1 expression levels was observed among nine different CD34+ subsets (28 and 27 samples from BM and CB, respectively) under the present experimental conditions, where detection limit of the WT1 expression was at 10−2 levels.

Table 1.

Expression of WT1 mRNA in FACS-Sorted Cells

BM CB 
Donor No. CD34+CD33 CD34+CD33+ Donor No. CD34+CD33 CD34+CD33+ 
  
      
 
2.0 × 10−2 <10−2 18 <10−2 <10−2 
<10−2 <10−2 19 <10−2 <10−2 
<10−2 <10−2 20 <10−2 <10−2 
<10−2 <10−2 21 1.5 × 10−2 <10−2 
<10−2 <10−2 
<10−2 <10−2 
BM CB 
Donor No. CD34+CD33 CD34+CD33+ Donor No. CD34+CD33 CD34+CD33+ 
  
      
 
2.0 × 10−2 <10−2 18 <10−2 <10−2 
<10−2 <10−2 19 <10−2 <10−2 
<10−2 <10−2 20 <10−2 <10−2 
<10−2 <10−2 21 1.5 × 10−2 <10−2 
<10−2 <10−2 
<10−2 <10−2 
Donor No. CD34+CD38 CD34+CD38+ Donor No. CD34+CD38 CD34+CD38+ 
  
      
 
<10−2 <10−2 22 <10−2 <10−2 
<10−2 1.3 × 10−2 23 <10−2 1.0 × 10−2 
   24 1.3 × 10−2 <10−2 
   25 1.7 × 10−2 1.0 × 10−2 
Donor No. CD34+CD38 CD34+CD38+ Donor No. CD34+CD38 CD34+CD38+ 
  
      
 
<10−2 <10−2 22 <10−2 <10−2 
<10−2 1.3 × 10−2 23 <10−2 1.0 × 10−2 
   24 1.3 × 10−2 <10−2 
   25 1.7 × 10−2 1.0 × 10−2 
Donor No. CD34+HLA-DR CD34+HLA-DR+ Donor No. CD34+HLA-DR CD34+HLA-DR+ 
  
      
 
1.3 × 10−2 2.2 × 10−2 26 NE <10−2 
10 NE 2.4 × 10−2 27 <10−2 1.0 × 10−2 
   28 1.4 × 10−2 2.4 × 10−2 
Donor No. CD34+HLA-DR CD34+HLA-DR+ Donor No. CD34+HLA-DR CD34+HLA-DR+ 
  
      
 
1.3 × 10−2 2.2 × 10−2 26 NE <10−2 
10 NE 2.4 × 10−2 27 <10−2 1.0 × 10−2 
   28 1.4 × 10−2 2.4 × 10−2 
Donor No. CD34+c-kithigh CD34+c-kitlow CD34+c-kit Donor No. CD34+c-kithigh CD34+c-kitlow CD34+c-kit 
  
        
 
11 <10−2 <10−2 <10−2 29 <10−2 <10−2 <10−2 
12 <10−2 <10−2 NE 30 <10−2 <10−2 1.0 × 10−2 
13 1.3 × 10−2 <10−2 NE 
14 1.0 × 10−2 1.5 × 10−2 NE 
Donor No. CD34+c-kithigh CD34+c-kitlow CD34+c-kit Donor No. CD34+c-kithigh CD34+c-kitlow CD34+c-kit 
  
        
 
11 <10−2 <10−2 <10−2 29 <10−2 <10−2 <10−2 
12 <10−2 <10−2 NE 30 <10−2 <10−2 1.0 × 10−2 
13 1.3 × 10−2 <10−2 NE 
14 1.0 × 10−2 1.5 × 10−2 NE 
Donor No. CD34+ CD34 Donor No. CD34+ CD34 
  
      
 
15 <10−2 <10−2 31 1.5 × 10−2 <10−2 
16 1.0 × 10−2 <10−2 32 2.4 × 10−2 <10−2 
17 2.2 × 10−2 <10−2 
Donor No. CD34+ CD34 Donor No. CD34+ CD34 
  
      
 
15 <10−2 <10−2 31 1.5 × 10−2 <10−2 
16 1.0 × 10−2 <10−2 32 2.4 × 10−2 <10−2 
17 2.2 × 10−2 <10−2 
FACS-Sorted Leukemic Cells
Patient No. FABCD34 (%)CD33 (%)UnsortedCD34+CD33+CD34+CD33CD34CD33+CD34CD33
293 68 58 1.5 × 10−1 5.4 × 10−1 4.8 × 10−1 2.9 × 10−1 1.6 × 10−1 
M1 
273 79 92 8.9 × 10−1 9.9 × 10−1 1.9 × 100  1.8 × 100  5.6 × 10−1 
M2 
194 10 72 6.4 × 10−1 3.4 × 10−1 8.9 × 10−1 3.6 × 10−1 8.6 × 10−1 
M4 
294 78 96 4.1 × 10−1 3.5 × 10−1 4.4 × 10−1 2.3 × 10−1 5.4 × 10−1 
M4 
271 91 1.0 × 10−2 ND ND 7.0 × 10−2 1.3 × 10−1 
M5b 
FACS-Sorted Leukemic Cells
Patient No. FABCD34 (%)CD33 (%)UnsortedCD34+CD33+CD34+CD33CD34CD33+CD34CD33
293 68 58 1.5 × 10−1 5.4 × 10−1 4.8 × 10−1 2.9 × 10−1 1.6 × 10−1 
M1 
273 79 92 8.9 × 10−1 9.9 × 10−1 1.9 × 100  1.8 × 100  5.6 × 10−1 
M2 
194 10 72 6.4 × 10−1 3.4 × 10−1 8.9 × 10−1 3.6 × 10−1 8.6 × 10−1 
M4 
294 78 96 4.1 × 10−1 3.5 × 10−1 4.4 × 10−1 2.3 × 10−1 5.4 × 10−1 
M4 
271 91 1.0 × 10−2 ND ND 7.0 × 10−2 1.3 × 10−1 
M5b 

Abbreviations: NE, not able to evaluate because of low levels of β-actin; ND, not determined; FAB, French-American-British criteria.

High levels of WT1 expression in leukemic cells.Fresh leukemic cells from 25 AML, 10 ALL, and 1 acute mixed-lineage leukemia (AMLL) patients were FACS-analyzed for the expression of CD34, CD33, CD38, and HLA-DR and examined for the expression levels of the WT1 gene (Table 2). WT1 expression levels of individual leukemic cell samples were plotted according to the levels of expression of CD34, CD33, CD38, and HLA-DR, which were either expressed or not expressed in the majority (>80%) of the leukemic cells (Fig 3). The average WT1 expression levels in leukemic cells were 2.4 to 9.3 × 10−1 and at least 10 times higher than those in hematopoietic progenitor cells of BM and CB, regardless of the expression of CD34, CD33, CD38, or HLA-DR. Next, fresh leukemic cells from 5 patients with AML were FACS-sorted according to the expression levels of CD34 and CD33 (Fig 1C). The average levels of WT1 expression in FACS-sorted CD34+CD33, CD34+CD33+, CD34CD33+, and CD34CD33 leukemic cell populations from 5 AML samples were 9.3 × 10−1, 5.6 × 10−1, 5.5 × 10−1, and 4.5 × 10−1, respectively (Figs 2 and 3 and Table 1). All of the four sorted leukemic cell populations expressed high levels of WT1, reflecting the phenotypical heterogeneity of leukemic cells. Moreover, various leukemic cell lines were examined for cell surface markers and WT1 expression (Fig 3 and Table 2). The WT1 expression levels in all but four leukemic cell lines examined (Molt-3, Tall-1, Jurkat, and Ball-1) were at least 13 times higher than those in hematopoietic progenitor cells of BM and CB, regardless of the expression of CD34, CD33, CD38, HLA-DR, and c-kit. Finally, fresh leukemic cells were used to analyze whether WT1 expression levels are related to the expression of CD34, CD33, CD38, and HLA-DR. WT1 expression levels were found to be significantly higher in CD34+HLA-DR than in CD34+HLA-DR+ leukemic cells (P < .001), and in CD34CD33+ than in CD34CD33 leukemic cells (P < .05).

Table 2.

Correlation Between WT1 Gene Expression Levels and Surface Markers

Leukemic Samples
FABPatient No.WT1 mRNACD34CD33CD38HLA-DR
L1 289 0.93 − − 
M1 80 0.57 − − 
AMLL 51 0.44 − 
ALL 81 0.34 − ND 
ALL 197 0.25 − − 
M2 124 0.11 − ND 
M2 237 0.074 − 
M2 220 0.022 − ND 
L2 234 0.021 − ND 
L1 275 0.012 − − 
M2 273 0.89 ND − 
M3 228 0.84 − − 
L2 139 0.70 ND 
M1 0.45 ND 
M4 294 0.41 
MDS-AML 318 0.30 ND 
MDS-AML 243 0.20 ND 
M4 227 0.05 ND 
ALL 288 0.026 ND 
M2 199 0.026 
M5a 37 1.72 − ND 
M1 317 1.34 − − 
M3 282 1.01 − ND − 
M6 287 0.98 − 
M4 275 0.97 − 
M5a 306 0.62 − 
M3 78 0.58 − ND − 
M2 235 0.45 − − 
M5 296 0.11 − 
M3 137 0.025 − ND − 
M5 274 0.013 − 
M5b 271 0.010 − ND 
L2 0.40 − − − 
ALL 280 0.25 − − ND 
M5a 33 0.050 − − 
L2 151 0.017 − − 
Leukemic Samples
FABPatient No.WT1 mRNACD34CD33CD38HLA-DR
L1 289 0.93 − − 
M1 80 0.57 − − 
AMLL 51 0.44 − 
ALL 81 0.34 − ND 
ALL 197 0.25 − − 
M2 124 0.11 − ND 
M2 237 0.074 − 
M2 220 0.022 − ND 
L2 234 0.021 − ND 
L1 275 0.012 − − 
M2 273 0.89 ND − 
M3 228 0.84 − − 
L2 139 0.70 ND 
M1 0.45 ND 
M4 294 0.41 
MDS-AML 318 0.30 ND 
MDS-AML 243 0.20 ND 
M4 227 0.05 ND 
ALL 288 0.026 ND 
M2 199 0.026 
M5a 37 1.72 − ND 
M1 317 1.34 − − 
M3 282 1.01 − ND − 
M6 287 0.98 − 
M4 275 0.97 − 
M5a 306 0.62 − 
M3 78 0.58 − ND − 
M2 235 0.45 − − 
M5 296 0.11 − 
M3 137 0.025 − ND − 
M5 274 0.013 − 
M5b 271 0.010 − ND 
L2 0.40 − − − 
ALL 280 0.25 − − ND 
M5a 33 0.050 − − 
L2 151 0.017 − − 
Leukemia and Lymphoma Cell Lines
Cell LineOriginWT1 mRNACD34CD33CD38HLA-DRc-kit
Molt-4 T-ALL 0.41 − − − 
KG-1 M1 1.12 − − +low 
KU812 CML 0.83 − +low 
TF-1 M6 0.80 +low 
Kasumi-1 M2 0.60 +low 
BV173 CML 1.50 − − 
K562 CML 1.00 − − − − 
HL-60 M3 0.56 − − − 
THP-1 M5 0.45 − − − 
HEL M6 0.37 − − +low 
IMS-M1 M5 0.31 − − − 
SAM-1 CML 0.71 − − − − − 
Molt-3 T-ALL 0.10 − − − − 
Tall-1 T-ALL 0.04 − − − − 
Jurkat T-ALL 2.0 × 10−4 − − − − 
Ball-1 B-ALL <10−5 − − ND 
Daudi Lymphoma 3.8 × 10−4 − − − 
U937 Lymphoma <10−5 − − − 
KT-3 Lymphoma <10−5 − − − − 
DL-40 Lymphoma <10−5 − − − 
Leukemia and Lymphoma Cell Lines
Cell LineOriginWT1 mRNACD34CD33CD38HLA-DRc-kit
Molt-4 T-ALL 0.41 − − − 
KG-1 M1 1.12 − − +low 
KU812 CML 0.83 − +low 
TF-1 M6 0.80 +low 
Kasumi-1 M2 0.60 +low 
BV173 CML 1.50 − − 
K562 CML 1.00 − − − − 
HL-60 M3 0.56 − − − 
THP-1 M5 0.45 − − − 
HEL M6 0.37 − − +low 
IMS-M1 M5 0.31 − − − 
SAM-1 CML 0.71 − − − − − 
Molt-3 T-ALL 0.10 − − − − 
Tall-1 T-ALL 0.04 − − − − 
Jurkat T-ALL 2.0 × 10−4 − − − − 
Ball-1 B-ALL <10−5 − − ND 
Daudi Lymphoma 3.8 × 10−4 − − − 
U937 Lymphoma <10−5 − − − 
KT-3 Lymphoma <10−5 − − − − 
DL-40 Lymphoma <10−5 − − − 

Abbreviation: ND, not determined; FAB, French-American-British criteria.

The WT1 expression in both the most primitive cells (CD34+CD33,11-17 CD34+CD38,18-20 CD34+HLA-DR,21-27 or CD34+c-kitlow 9,28-35) and the more mature progenitor cells (CD34+CD33+, CD34+CD38+, CD34+HLA-DR+, or CD34+c-kithigh) of BM and CB was identified at very low (1.0 to 2.4 × 10−2) or undetectable (<10−2) levels. Moreover, the WT1 expression in more differentiated CD34 cells was undetectable in all of the 5 CD34 cell samples. However, these results cannot exclude the possibility of a transient expression of WT1 among a subpopulation of hematopoietic precursors. On the other hand, fresh leukemic cells expressed high levels (average, 2.4 to 9.3 × 10−1) of WT1, regardless of cell surface markers. Thus, the WT1 expression levels in normal hematopoietic progenitor cells were at least one log less than those in leukemic cells. Before we obtained the present data, we assumed that we could easily find normal counterparts for the leukemic cells with comparable WT1 expression levels; however, we could not. This suggests that overexpression of the WT1 gene in leukemic cells reflects a leukemic state of hematopoietic progenitor cells, but not WT1 expression levels in normal counterparts.

Fraizer et al36 have recently reported that CD34+ BM cells express WT1 at levels comparable to those in K562 and fresh leukemic cells. Their results are not compatible with ours in that the expression levels of WT1 not only in the CD34+and CD34 cell populations, but also in the CD34+CD33, CD34+CD33+, CD34+CD38, CD34+CD38+, CD34+HLA-DR, CD34+HLA-DR+, CD34+c-kithigh, CD34+c-kitlow, and CD34+c-kit subsets were very low (1.0 to 2.4 × 10−2) or undetectable (<10−2) and at least 10 times less than those in leukemic cells. The precise cause of this discrepancy between their results and ours is unknown. However, it might be ascribed to the difference in the optimal conditions for RT-PCR (our RT-PCR conditions were optimized as described here and previously5,6 ). Menssen et al37 used an immunofluorescence assay using the anti-WT1 MoAb and found the WT1 protein only in the nuclei of leukemic blast cells, but not in the nuclei of normal CD34+ hematopoietic progenitor cells. Moreover, the WT1 transcript was detected by RT-PCR assay in 68 of 86 (79%) ALL patients and in 53 of 57 (93%) AML patients. In contrast, normal peripheral CD34+ hematopoietic progenitor cells did not express the WT1 gene at detectable levels. Thus, the data presented here are basically compatible with the findings of Menssen et al, although the detection limit (1 in 5,000) for WT1 transcript of their RT-PCR assay is 20 times lower than that (1 in 100,000) of our RT-PCR assay, and it remains unclear whether peripheral CD34+ cells can be compared with the CD34+ cells of BM and CB.

Of 5 CD34+ cell samples, 4 expressed very low levels (1.0 to 2.4 × 10−2) of WT1, whereas the WT1 expression levels in all of the 5 CD34 cell samples were undetectable (<10−2). Thus, the WT1 expression levels were significantly (P < .01) higher in CD34+ cells than in CD34 cells, suggesting that the WT1 expression levels decrease as hematopoietic progenitor cells differentiate into more mature cells. Moreover, the WT1 expression levels in the CD34+HLA-DR and CD34CD33+ leukemic cells were respectively and significantly higher than those in the more differentiated CD34+HLA-DR+ and CD34CD33 leukemic cells. The WT1 expression levels in mature T- and B-cell ALL cell lines (Molt-3, Tall-1, Jurkat, and Ball-1) were very low. This is consistent with the findings that the WT1 expression was downregulated during the differentiation of HL60 cells by dimethyl sulfoxide or retinoic acid38 and during induction of erythroid and megakaryocytic differentiation of K562 cells.39 

When leukemic cell samples from 15 acute leukemia patients were subjected to PCR single-strand conformation polymorphism (SSCP) analysis, no point mutations were found within the zinc finger region.5 Furthermore, when 12 of 15 leukemia samples were examined by SSCP analysis for point mutations within the region (exons 2 to 6) 5′ to the zinc finger region, no point mutations were detected.5 On the other hand, King-Underwood et al40 have recently reported that they have found mutations in the WT1 gene in 4 of 36 acute leukemia samples by SSCP analysis. The mutations comprise small insertions in exons 1 and 7 and a nonsense mutation in exon 9, predicting the production of a truncated WT1 protein with absence or disruption of the zinc finger region. Determination of biological significance of WT1 mutations should clarify the roles of the WT1 gene in leukemogenesis.

The mechanism of overexpression of the WT1 gene in human leukemic cells remains undetermined. Recently, Wu et al41 characterized the human WT1 enhancer region located at the 3′ of the WT1 gene, and showed that the activity of the 3′ WT1 enhancer was positively regulated by erythrocyte/megakaryocyte-related transcription factor GATA-1. Shimamoto et al42 reported that GATA-1 was expressed in 30 of 62 AML, 2 of 19 ALL, and 9 of 14 CML blastic crisis. Thus, GATA-1 expression might play a part in WT1 overexpression in leukemia. However, GATA-1 expression is limited to a part of leukemia, whereas WT1 overexpression is general in leukemia. Therefore, as yet undetermined mechanisms should play an important role in an aberrant overexpression of WT1 in leukemia.

Fresh leukemic cells were FACS-sorted and fractionized into four populations (CD34+CD33, CD34+CD33+, CD34CD33+, and CD34CD33) according to the expression levels of CD34 and CD33. Interestingly, all four populations expressed high levels of WT1, suggesting that these four populations all consisted of leukemic cells and also that these leukemic cells were phenotypically heterogenous. These results indicate that a leukemic clone that was generated by malignant transformation of a hematopoietic progenitor cell became heterogenous during its proliferation and expansion.

Mononuclear cells of normal BM expressed 10−4 levels of WT1.5 It remains unclear which cells are responsible for these background levels of WT1 expression, because the present study could not definitively identify such cells. However, because low levels of WT1 (10−2 levels) were expressed in 4 of the 5 CD34+ cell samples and in 17 of the 55 CD34+ subsets, whereas in all of the 5 CD34 cell samples WT1 expression was at undetectable levels (<10−2), CD34+ cells appear to be responsible for the background levels of WT1 expression in BM.

We thank Tsuyomi Noguchi and Hiromi Takeuchi for typing this manuscript and Machiko Mishima for her skillful technical assistance with the PCR.

Address reprint requests to Haruo Sugiyama, MD, Departments of Clinical Laboratory Science and Medicine III, Osaka University Medical School, 2-2, Yamada-Oka, Suita City, Osaka 565, Japan.

1
Buckler
AJ
Pelletier
J
Haber
DA
Glaser
T
Housman
DE
Isolation, characterization, and expression of the murine Wilms' tumor gene (WT1) during kidney development.
Mol Cell Biol
11
1991
1707
2
Park
S
Schalling
M
Bernard
A
Maheswaran
S
Shipley
GC
Roberts
D
Fletcher
J
Shipman
R
Rheinwald
J
Demetri
G
Griffin
J
Minden
M
Housman
DE
Haber
DA
The Wilms tumor gene WT1 is expressed in murine mesoderm-derived tissues and mutated in a human mesothelioma.
Nat Genet
4
1993
415
3
Kreidberg
JA
Sariola
H
Loring
JM
Maeda
M
Pelletier
J
Housman
D
Jaenisch
R
WT-1 is required for early kidney development.
Cell
74
1993
679
4
Miwa
H
Beran
M
Saunders
GF
Expression of the Wilms tumor gene (WT1) in human leukemias.
Leukemia
6
1992
405
5
Inoue
K
Sugiyama
H
Ogawa
H
Nakagawa
M
Yamagami
T
Miwa
H
Kita
K
Hiraoka
A
Masaoka
T
Nasu
K
Kyo
T
Dohy
H
Nakauchi
H
Ishidate
T
Akiyama
T
Kishimoto
T
WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia.
Blood
84
1994
3071
6
Inoue
K
Ogawa
H
Yamagami
T
Soma
T
Tani
Y
Tatekawa
T
Oji
Y
Tamaki
H
Kyo
T
Dohy
H
Hiraoka
A
Masaoka
T
Kishimoto
T
Sugiyama
H
Long-term follow-up of minimal residual disease in leukemia patients by monitoring WT1 (Wilms tumor gene) expression levels.
Blood
88
1996
2267
7
Yamagami
T
Sugiyama
H
Inoue
K
Ogawa
H
Tatekawa
T
Hirata
M
Kudoh
T
Akiyama
T
Murakami
A
Maekawa
T
Kishimoto
T
Growth inhibition of human leukemic cells by WT1 (Wilms tumor gene) antisense oligodeoxynucleotides: Implications for the involvement of WT1 in leukemogenesis.
Blood
87
1996
2878
8
Sonoda
Y
Kuzuyama
T
Tanaka
S
Yokota
S
Maekawa
T
Clark
SC
Abe
T
Human interleukin-4 inhibits proliferation of megakaryocyte progenitor cells in culture.
Blood
81
1993
624
9
Sonoda
Y
Sakabe
H
Ohmisono
Y
Tanimukai
S
Yokota
S
Nakagawa
S
Clark
SC
Abe
T
Synergistic actions of stem cell factor and other burst-promoting activities on proliferation of CD34+ highly purified blood progenitors expressing HLA-DR or different levels of c-kit protein.
Blood
84
1994
4099
10
Inoue
K
Sugiyama
H
Ogawa
H
Yamagami
T
Azuma
T
Oka
Y
Miwa
H
Kita
K
Hiraoka
A
Masaoka
T
Nasu
K
Kyo
T
Dohy
H
Hara
J
Kanamaru
A
Kishimoto
T
Expression of the interleukin- 6 (IL-6), IL-6 receptor, and gp130 genes in acute leukemia.
Blood
84
1994
2672
11
Ema
H
Suda
T
Miura
Y
Nakauchi
H
Colony formation of clone-sorted human hematopoietic progenitors.
Blood
75
1990
1941
12
Bernstein
ID
Leary
AG
Andrews
RG
Ogawa
M
Blast colony-forming cells and precursors of colony-forming cells detectable in long-term marrow culture express the same phenotype (CD33− CD34+).
Exp Hematol
19
1991
680
13
Bender
JG
Unverzagt
KL
Walker
DE
Lee
W
Van Epps
DE
Smith
DH
Stewart
CC
To
LB
Identification and comparison of CD34-positive cells and their subpopulations from normal peripheral blood and bone marrow using multicolor flow cytometry.
Blood
77
1991
2591
14
Serke
S
Säuberlich
S
Abe
Y
Huhn
D
Analysis of CD34-positive hemopoietic progenitor cells from normal human adult peripheral blood: Flow-cytometrical studies and in-vitro colony (CFU-GM, BFU-E) assays.
Ann Hematol
62
1991
45
15
Saeland
S
Duvert
V
Caux
C
Pandrau
D
Favre
C
Vallé
A
Durand
I
Charbord
P
de Vries
J
Banchereau
J
Distribution of surface-membrane molecules on bone marrow and cord blood CD34+ hematopoietic cells.
Exp Hematol
20
1992
24
16
Kinniburgh
D
Russell
NH
Comparative study of CD34-positive cells and subpopulations in human umbilical cord blood and bone marrow.
Bone Marrow Transplant
12
1993
489
17
Thoma
SJ
Lamping
CP
Ziegler
BL
Phenotype analysis of hemaotopoietic CD34+ cell populations derived from human umbilical cord blood using flow cytometry and cDNA-polymerase chain reaction.
Blood
83
1994
2103
18
Terstappen
LWMM
Huan
S
Safford
M
Lansdorp
PM
Loken
MR
Sequential generations of hematopoietic colonies derived from single nonlineage-committed CD34+CD38− progenitor cells.
Blood
77
1991
1218
19
Terstappen
LWMM
Buescher
S
Nguyen
M
Reading
C
Differentiation and maturation of growth factor expanded human hematopoietic progenitors assessed by multidimensional flow cytometry.
Leukemia
6
1992
1001
20
Issaad
C
Croisille
L
Katz
A
Vainchenker
W
Coulombel
L
A murine stromal cell line allows the proliferation of very primitive human CD34++/CD38− progenitor cells in long-term cultures and semisolid assays.
Blood
81
1993
2916
21
Moore
MAS
Broxmeyer
HE
Sheridan
APC
Meyers
PA
Jacobsen
N
Winchester
RJ
Continuous human bone marrow culture: Ia antigen characterization of probable pluripotential stem cells.
Blood
55
1980
682
22
Keating
A
Powell
J
Takahashi
M
Singer
JW
The generation of human long-term marrow cultures from marrow depleted of Ia (HLA-DR) positive cells.
Blood
64
1984
1159
23
Brandt
J
Baird
N
Lu
L
Srour
E
Hoffman
R
Characterization of a human hematopoietic progenitor cell capable of forming blast cell containing colonies in vitro.
J Clin Invest
82
1988
1017
24
Sutherland
HJ
Eaves
CJ
Eaves
AC
Dragowska
W
Lansdorp
PM
Charcterization and partial purification of human marrow cells capable of initiating long-term hematopoiesis in vitro.
Blood
74
1989
1563
25
Caux
C
Favre
C
Saeland
S
Duvert
V
Mannoni
P
Durand
I
Aubry
J-P
de Vries
J
Sequential loss of CD34 and class II MHC antigens on purified cord blood hematopoietic progenitors cultured with IL-3: Characterization of CD34−, HLA-DR+ cells.
Blood
74
1989
1287
26
Verfaillie
C
Blakolmer
K
McGlave
P
Purified primitive human hematopoietic progenitor cells with long-term in vitro repopulating capacity adhere selectively to irradiated bone marrow stroma.
J Exp Med
172
1990
509
27
Udomsakdi
C
Lansdorp
PM
Hogge
DE
Reid
DS
Eaves
AC
Eaves
CJ
Characterization of primitive hematopoietic cells in normal human peripheral blood.
Blood
80
1992
2513
28
Ogawa
M
Matsuzaki
Y
Nishikawa
S
Hayashi
S-I
Kunisada
T
Sudo
T
Kina
T
Nakauchi
H
Nishikawa
S-I
Expression and function of c-kit in hematopoietic progenitor cells.
J Exp Med
174
1991
63
29
Catlett
JP
Leftwich
JA
Westin
EH
Grant
S
Huff
TF
c-kit expression by CD34+ bone marrow progenitors and inhibition of response to recombinant human interleukin-3 following exposure to c-kit anitsense oligonucleotides.
Blood
78
1991
3186
30
Papayannopoulou
T
Brice
M
Broudy
VC
Zsebo
KM
Isolation of c-kit receptor-expressing cells from bone marrow, peripheral blood, and fetal liver: Functional properties and composite antigenic profile.
Blood
78
1991
1403
31
Ratajczak
MZ
Luger
SM
DeRiel
K
Abrahm
J
Calabretta
B
Gewirtz
AM
Role of the KIT protooncogene in normal and malignant human hematopoiesis.
Proc Natl Acad Sci USA
89
1992
1710
32
Briddell
RA
Broudy
VC
Bruno
E
Brandt
JE
Srour
EF
Hoffman
R
Further phenotypic characterization and isolation of human hematopoietic progenitor cells using a monoclonal antibody to the c-kit receptor.
Blood
79
1992
3159
33
Orlic
D
Fischer
R
Nishikawa
S-I
Nienhuis
AW
Bodine
DM
Purification and characterization of heterogeneous pluripotent hematopoietic stem cell populations expressing high levels of c-kit receptor.
Blood
82
1993
762
34
Gunji
Y
Nakamura
M
Osawa
H
Nagayoshi
K
Nakauchi
H
Miura
Y
Yanagisawa
M
Suda
T
Human primitive hematopoietic progenitor cells are more enriched in KITlow cells than in KIThigh cells.
Blood
82
1993
3283
35
Laver
JH
Abboud
MR
Kawashima
I
Leary
AG
Ashman
LK
Ogawa
M
Characterization of c-kit expression by primitive hematopoietic progenitors in umbilical cord blood.
Exp Hematol
23
1995
1515
36
Fraizer
GC
Patmasiriwat
P
Zhang
X
Saunders
GF
Expression of the tumor suppressor gene WT1 in both human and mouse bone marrow.
Blood
86
1995
4704
37
Menssen
HD
Renkl
H-J
Rodeck
U
Maurer
J
Notter
M
Schwartz
S
Reinhardt
R
Thiel
E
Presence of Wilms' tumor gene (WT1) transcripts and the WT1 nuclear protein in the majority of human acute leukemias.
Leukemia
9
1995
1060
38
Sekiya
M
Adachi
M
Hinoda
Y
Imai
K
Yachi
A
Downregulation of Wilms' tumor gene (WT1) during myelomonocytic differentiation in HL60 cells.
Blood
83
1994
1876
39
Phelan
SA
Lindberg
C
Call
KM
Wilms' tumor gene, WT1, mRNA is down-regulated during induction of erythroid and megakaryocytic differentiation of K562 cells.
Cell Growth Differ
5
1994
677
40
King-Underwood
L
Renshaw
J
Pritchard-Jones
K
Mutations in the Wilms' tumor gene WT1 in leukemias.
Blood
87
1996
2171
41
Wu
Y-J
Fraizer
GC
Saunders
GF
GATA-1 transactivates the WT1 hematopoietic specific enhancer.
J Biol Chem
270
1995
5944
42
Shimamoto
T
Ohyashiki
K
Ohyashiki
JH
Kawakubo
K
Fujimura
T
Iwama
H
Nakazawa
S
Toyama
K
The expression pattern of erythrocyte/megakaryocyte-related transcription factors GATA-1 and the stem cell leukemia gene correlates with hematopoietic differentiation and is associated with outcome of acute myeloid leukemia.
Blood
86
1995
3173
Sign in via your Institution