Accepted Manuscript ESAM is a novel human hematopoietic stem cell marker associated with a subset of human leukemias Tomohiko Ishibashi, Takafumi Yokota, Hirokazu Tanaka, Michiko Ichii, Takao Sudo, Yusuke Satoh, Yukiko Doi, Tomoaki Ueda, Akira Tanimura, Yuri Hamanaka, Sachiko Ezoe, Hirohiko Shibayama, Kenji Oritani, Yuzuru Kanakura PII:
S0301-472X(16)00005-9
DOI:
10.1016/j.exphem.2015.12.010
Reference:
EXPHEM 3351
To appear in:
Experimental Hematology
Received Date: 14 January 2015 Revised Date:
24 December 2015
Accepted Date: 28 December 2015
Please cite this article as: Ishibashi T, Yokota T, Tanaka H, Ichii M, Sudo T, Satoh Y, Doi Y, Ueda T, Tanimura A, Hamanaka Y, Ezoe S, Shibayama H, Oritani K, Kanakura Y, ESAM is a novel human hematopoietic stem cell marker associated with a subset of human leukemias, Experimental Hematology (2016), doi: 10.1016/j.exphem.2015.12.010. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT 1
ESAM is a novel human hematopoietic stem cell marker associated with a subset of
2
human leukemias
3 Authors
5
Tomohiko Ishibashi1, Takafumi Yokota1, Hirokazu Tanaka2, Michiko Ichii1,
6
Takao Sudo1, Yusuke Satoh1,3, Yukiko Doi1, Tomoaki Ueda1, Akira Tanimura1,
7
Yuri Hamanaka1, Sachiko Ezoe1, Hirohiko Shibayama1, Kenji Oritani1, Yuzuru Kanakura1
SC
RI PT
4
8 Affiliations
10
1
11
Suita, Osaka 565-0871, Japan
12
2
13
Osakasayama, Osaka 589-8511, Japan
14
3
M AN U
9
Department of Hematology and Oncology, Osaka University Graduate School of Medicine,
TE D
Department of Hematology and Rheumatology, Kinki University Faculty of Medicine,
Department of Lifestyle Studies, Kobe Shoin Women’s University, Kobe 657-0015, Japan
15 Contact:
17
Takafumi Yokota, Department of Hematology and Oncology, Osaka University Graduate
18
School of Medicine, C9, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; E-mail:
19
[email protected]
AC C
20
EP
16
21
Category for the Table of Contents
22
Stem Cells (hematopoietic, mesenchymal, embryonic and induced pluripotent stem cells)
23 24
Word Count: 3,530 words
1
ACCEPTED MANUSCRIPT 25 26
Conflict of interest
27
The authors declare no conflict of interest.
AC C
EP
TE D
M AN U
SC
RI PT
28
2
ACCEPTED MANUSCRIPT Abstract
30
Reliable markers are essential to increase our understanding of the biological features of
31
human hematopoietic stem cells and to facilitate the application of hematopoietic stem cells
32
in the field of transplantation and regenerative medicine. We previously identified endothelial
33
cell-selective adhesion molecule (ESAM) as a novel functional marker of hematopoietic stem
34
cells in mice. Here, we found that ESAM can also be used to purify human hematopoietic
35
stem cells from all the currently available sources (adult bone marrow, mobilized peripheral
36
blood, and cord blood). Multipotent colony-forming units and long-term
37
hematopoietic-reconstituting cells in immunodeficient mice were exclusively found in the
38
ESAMHigh fraction of CD34+ CD38– cells. The CD34+ CD38– fraction of cord blood and
39
collagenase-treated bone marrow contained cells showing extremely high expression of
40
ESAM; these cells are likely to be related to the endothelial lineage. Leukemia cell lines of
41
erythroid and megakaryocyte origin, but not those of myeloid or lymphoid descent, were
42
ESAM-positive. However, high ESAM expression was observed in some primary acute
43
myeloid leukemia cells. Furthermore, KG-1a myeloid leukemia cells switched from
44
ESAM-negative to ESAM-positive with repeated leukemia reconstitution in vivo. Thus,
45
ESAM is a useful marker for studying both human hematopoietic stem cells and leukemia
46
cells.
48
SC
M AN U
TE D
EP
AC C
47
RI PT
29
3
ACCEPTED MANUSCRIPT 49
Introduction
50
Lifelong hematopoiesis is maintained by hematopoietic stem cells (HSCs), which have self-renewal capacities and can differentiate into all types of blood cells. HSCs are
52
essential for transplantation and regenerative therapies designed to cure patients with
53
hematological diseases. The successful outcome of therapy requires a reliable method for
54
identifying authentic HSCs. Therefore, it is important to identify cell surface markers that
55
distinguish HSCs from other cells.
SC
RI PT
51
56
Since a previous study showed that a set of cell surface proteins can be used to enrich multipotent hematopoietic progenitors in the mouse bone marrow (BM) [1], numerous
58
studies have examined murine HSC markers (reviewed by Yokota et al. [2]). Murine HSCs
59
can be efficiently isolated from BM as Lin– Sca-1+ c-kit+ (LSK) CD90/Thy1Low CD34–/Low
60
CD150+ CD48– cells. Indeed, ~50% of cells with the LSK CD150+ CD48– phenotype exhibit
61
the potential for long-term multi-lineage reconstitution [3]. In humans, however, the same
62
method cannot be applied because of critical differences between the murine and human HSC
63
phenotypes. Human HSCs express neither CD150 nor Sca-1 [4, 5]. In addition, human HSCs
64
are enriched in the CD34+ CD38– fraction [6, 7], whereas many studies have located murine
65
HSCs in the CD34– CD38+ fraction [8-10]. These discrepancies have limited the translation
66
of the findings from mice to humans. Therefore, the identification of common HSC antigens
67
between the two species would be a significant advancement in translational studies of HSC
68
biology.
TE D
EP
AC C
69
M AN U
57
We previously identified endothelial cell-selective adhesion molecule (ESAM) as a
70
novel marker for murine HSCs [11]. The functional importance of ESAM is underscored by
71
the finding that ESAM-deficient mice suffer from severe and prolonged BM suppression after
72
treatment with the cytotoxic agent 5-fluorouracil (5-FU) [12]. The detection of ESAM
4
ACCEPTED MANUSCRIPT transcripts in human cord blood (CB) CD34+ CD38– Lin– CD90/Thy1+ cells has increased
74
interest in ESAM as an HSC marker [13]. However, it is unclear whether its expression
75
pattern changes in human hematopoietic stem/progenitor cells with the differentiation status,
76
or in a manner that depends on the tissue source. In addition, the expression of ESAM in a
77
broad panel of human leukemia cells has not been evaluated.
78
RI PT
73
In this study, we examined whether ESAM expression is a potential marker of human HSCs from diverse sources. Interestingly, in addition to adult BM (ABM), most
80
CD34+ CD38– cells in granulocyte-colony-stimulating factor (G-CSF)-mobilized peripheral
81
blood (GMPB) expressed ESAM. We also identified a previously uncharacterized subset of
82
CB CD34+ CD38– cells that expressed extremely high levels of ESAM. Furthermore, we
83
observed oscillations in ESAM expression on leukemia cells depending on the surrounding
84
environment.
M AN U
SC
79
86
Materials and methods
87
Human samples
BM and GMPB were obtained from related HSC transplantation donors. CB was
EP
88
TE D
85
obtained from healthy full-term newborns. BM was also obtained from patients of acute
90
leukemia, and the femoral head of patients who underwent hip replacement surgery. The
91
Institutional Review Board at the Osaka University Hospital approved all protocols, and
92
written informed consent was obtained from all the participants. To prepare BM cells
93
adjacent to bone tissues, trabecular tissues of the femora were treated with 2 mg/mL
94
collagenase IV and DNase and gently agitated for 1 h at 37°C.
AC C
89
95 96
Additional methods are described in detail in the Supplementary methods.
5
ACCEPTED MANUSCRIPT 97 98
Results
99 High ESAM expression indicates primitive hematopoietic progenitors in human CD34+
101
CD38– cells
RI PT
100
Our previous study demonstrated that sorting for high ESAM expression enriches
103
the proportion of authentic HSCs in the LSK fraction of murine fetal liver and ABM. In this
104
study, we examined whether this was also true for human cells. Using flow cytometry, we
105
analyzed ESAM expression on BM mononuclear cells (MNCs). A large number of human
106
BM CD34+ cells were found to express ESAM, and most CD34+ CD38– cells, which
107
included the human HSC-enriched fraction, expressed ESAM (Figure 1A). We found that in
108
both ABM and GMPB, more than 80% of CD34+ CD38– cells expressed high levels of
109
ESAM (Figure 1B, C). The addition of anti-CD90 and CD45RA antibodies can reportedly
110
divide the CD34+ CD38– fraction into three subpopulations, including CD90+ CD45RA– HSC,
111
CD90– CD45RA– multipotent progenitors, and CD90– CD45RA+ multi-lymphoid progenitors
112
(MLP) [14] (Figure 1D, left). We found that most HSCs expressed high levels of ESAM,
113
whereas many MLPs lost ESAM expression (Figure 1D, middle). Furthermore, estimation of
114
the mean fluorescence intensity indicated that ESAM expression markedly declined as the
115
cells progressively differentiated from HSCs into MLPs (Figure 1D, right).
M AN U
TE D
EP
AC C
116
SC
102
Next, we compared the differentiation and growth potential of ESAM–/Low and
117
ESAMHigh cells. We purified CD34+ CD38– ESAM–/Low and CD34+ CD38– ESAMHigh cells
118
from ABM, and subjected the cells to a methylcellulose assay or grew the cells in MS5
119
stromal cell co-culture. While both cell types produced hematopoietic colonies with high
120
frequency, only ESAMHigh cells included lineage-mixed colony-forming units (CFU-Mix)
6
ACCEPTED MANUSCRIPT with both myeloid and erythroid potential (Figure 2A). Under the MS5 co-culture conditions,
122
the ESAM–/Low cells generated CD45+ hematopoietic cells more effectively than the
123
ESAMHigh cells during the first week (Figure 2B, left); however, the output of CD45+ cells
124
did not increase to the same extent as that from ESAMHigh cells during the second week and
125
thereafter (Figure 2B, right). Indeed, hematopoietic cells rapidly expanded from the
126
ESAMHigh fraction after the second week, and this was maintained throughout the culture
127
period (Figure 2B). In addition, the ESAMHigh fraction gave rise to a substantial number of
128
CD19+ B-lineage cells (Figure 2C). Similar results were obtained when GMPB-derived cells
129
were used (data not shown). These observations indicate that while both the ESAM–/Low and
130
ESAMHigh cells in CD34+ CD38– included hematopoietic progenitors, higher ESAM
131
expression distinguished the more primitive and multipotent cells.
M AN U
SC
RI PT
121
132
High ESAM expression reveals a new subpopulation of human CB CD34+ CD38– cells
134
We also analyzed ESAM expression on human CB MNCs. Similarly to ABM and
TE D
133
GMPB, most cells in the CB CD34+ CD38– fraction expressed ESAM. However, we detected
136
a new sub-fraction of cells showing very high ESAM expression, referred to as ESAMBright,
137
which was not observed in the ABM and GMPB CD34+ CD38– fractions. The ESAMBright
138
fraction showed approximately 10-fold higher expression of ESAM compared to the
139
ESAMHigh fraction. According to the ESAM expression level, we subdivided the CB CD34+
140
CD38– fraction into three populations, including ESAM–/Low, ESAMHigh, and ESAMBright
141
(Figure 3A). While the percentage of ESAM–/Low cells among the total CD34+ CD38– cells
142
was consistently low in the tested CB samples, the frequencies of ESAMHigh and ESAMBright
143
cells varied widely among samples (Figure 3B).
AC C
EP
135
144
7
ACCEPTED MANUSCRIPT 145 146
ESAMHigh, but not ESAMBright, indicate authentic HSCs in CB Next, we performed functional assessment of the subpopulations based on ESAM expression levels. In some cases, ESAM–/Low cells were omitted from the assessment because
148
their frequency in the CB CD34+ CD38– fraction was too low for accurate evaluation. We
149
predicted that ESAMBright cells would be enriched for more primitive progenitors; however,
150
the opposite was observed. In methylcellulose assays, the ESAMHigh fraction produced
151
significantly more CFUs than the ESAMBright fraction (Figure 4A). Furthermore, only the
152
ESAMHigh fraction was capable of colony formation in the CFU-mix, which contained
153
primitive multipotent progenitors.
SC
M AN U
154
RI PT
147
To determine the frequency of primitive progenitors with both lymphoid and myeloid potential, limiting dilution analyses were performed in the MS5 co-culture system.
156
While 1 in 12 ESAMHigh cells produced both CD19+ lymphoid lineage and CD33+ myeloid
157
lineage cells, only 1 in 63 ESAMBright cells produced both lineage cells (Figure 4B). These
158
results suggest that primitive myelo-lymphoid progenitors are enriched in the ESAMHigh
159
fraction, but not in the ESAMBright population, of human CB CD34+ CD38– cells. To evaluate the long-term reconstitution activity, we first transplanted ESAMHigh or
EP
160
TE D
155
ESAMBright cells from the CB CD34+ CD38– fraction into 2.2 Gy-irradiated NOD/Shi-scid,
162
IL-2γ-null (NOG) mice. While all five recipients of the ESAMBright cells died within 1 month
163
after transplantation, all five recipients of the ESAMHigh cells survived. Surviving
164
ESAMHigh-transplanted mice were sacrificed 7 months after transplantation, and BM was
165
evaluated. We found that the ESAMHigh cells effectively reconstituted human hematopoiesis
166
in all recipients (data not shown). Next, we performed serial transplantation assays to
167
compare ESAM–/Low, ESAMHigh, and ESAMBright cells purified from the CB CD34+ CD38–
168
fraction. The number of transplanted cells in each group was determined according to their
AC C
161
8
ACCEPTED MANUSCRIPT frequency in the CB CD34+ CD38– fraction (see Figure 4 legend). Since all
170
ESAMBright-transplanted mice died before assessment in the first experiment, we reduced the
171
irradiation dose to 1.8 Gy. All recipients survived and were analyzed 3 months after
172
transplantation. Human hematopoiesis was only observed in mice transplanted with
173
ESAMHigh cells (Figure 4C). Furthermore, transplanted cells contributed substantially to the
174
myeloid, B-lymphoid, and T-lymphoid lineages (Figure 4D). In the secondary transplantation,
175
whole BM MNCs from the recipients of ESAMHigh cells were transplanted into sub-lethally
176
irradiated NOG mice. All mice were sacrificed 4 months after transplantation, and chimerism
177
of human cells in the BM was evaluated. Long-term human hematopoiesis was again
178
detected in the three lineages (Figure 4E and 4F). Thus, the ESAMHigh fraction, but not the
179
ESAM–/Low and ESAMBright fractions, contains long-term reconstituting human HSCs.
M AN U
SC
RI PT
169
180
182
CD34+ CD38– ESAMBright fraction consists of non-hematopoietic lineage cells
TE D
181
The unexpected results of our functional evaluation of CB CD34+ CD38– ESAMBright cells prompted us to characterize these cells more precisely. Morphologically,
184
ESAMBright cells were larger in size and had less basophilic and more abundant cytoplasm
185
compared with ESAM–/Low and ESAMHigh cells (Figure 5A). While ESAM–/Low and
186
ESAMHigh cells were of uniform size and morphology, ESAMBright cells were polymorphic,
187
suggesting that they are heterogeneous.
AC C
188
EP
183
With respect to the cell surface phenotype, ESAMBright cells did not express CD45
189
(Figure 5B, left). In addition, while ESAMHigh cells expressed CD133, another marker of
190
human HSCs [15, 16], this marker was completely absent in ESAMBright cells (Figure 5B,
191
right). Back plotting of the ESAMHigh and ESAMBright cells onto the CD34 and CD38 profile
192
showed that CD34 expression levels were much higher on ESAMBright cells (Figure 5C).
9
ACCEPTED MANUSCRIPT Furthermore, these cells expressed vascular endothelial (VE)-cadherin and vascular
194
endothelial growth factor receptor-2 (VEGFR-2), which are endothelial lineage-related
195
molecules (Figure 5D). ESAMBright cells also expressed CD146/S-endo, and a small subset
196
was positive for the expression of CD118/leukemia inhibitory factor receptor (data not
197
shown). These results suggest that the main population of CB CD34+ CD38– ESAMBright cells
198
was not hematopoietic but rather comprised of endothelial lineage and other mesodermal
199
cells.
SC
200
RI PT
193
Based on the above findings, we assumed that a similar ESAMBright cell population existed in BM. Since aspirated BM cells did not contain ESAMBright cells (Figure 1B), we
202
treated bone samples with collagenase to collect mesenchymal cells that are adherent to bone
203
tissues. This treatment yielded a substantial CD34+ CD38– ESAMBright population from ABM
204
(Figure 5E), the phenotype of which was similar to its CB counterpart. We then performed a
205
microarray experiment to compare the gene expression profiles of the CD34+ CD38–
206
ESAMHigh and CD34+ CD38– ESAMBright populations. The former cells expressed
207
HSC-related genes, whereas the latter showed more endothelial-related profiles [17] (Figure
208
5F; the data have been deposited in Gene Expression Omnibus under accession number
209
GSE63877). Indeed, CD34+ CD38– ESAMBright cells from collagenase-treated bone produced
210
CD31+ cells, but not CD45+ hematopoietic cells, in co-culture with MS5 stromal cells in the
211
presence of VEGF, stromal cell-derived factor, and interleukin (IL)-16 (Figure 5G). These
212
observations indicate that the CD34+ CD38– fraction, which is conventionally considered the
213
human HSC fraction, may also contain a substantial number of non-hematopoietic cells. Thus,
214
the inclusion of ESAM or other markers appears to provide a more accurate estimation of
215
HSC numbers.
AC C
EP
TE D
M AN U
201
216
10
ACCEPTED MANUSCRIPT 217 218
Some types of human leukemia cells express ESAM Cell surface antigens on normal hematopoietic stem/progenitor cells are often observed on leukemia cells. Since some of these antigens are useful for determining leukemia
220
lineage and have utility as prognostic indicators [18, 19], we examined whether ESAM would
221
be a valuable addition to this antigen panel. First, we examined human leukemia cell lines
222
derived from patients with acute myeloid leukemia (AML); all of these cell lines were
223
uniformly negative for ESAM expression (Figure 6A). KG-1a cells showed a contrasting
224
profile for CD34 and ESAM. Lymphoid lineage cell lines were also negative for ESAM. In
225
contrast, the erythroid leukemia cell line HEL and the megakaryocytic leukemia cell line
226
CMK showed high ESAM expression. Additionally, K562 cells that originated from a
227
chronic myeloid leukemia that subsequently transformed into acute erythroleukemia also
228
expressed low levels of ESAM. These results indicate that ESAM expression segregates
229
leukemia cells with erythroid or megakaryocyte lineage.
SC
M AN U
TE D
230
RI PT
219
We then evaluated ESAM expression on primary leukemia cells isolated from patients with AML (n = 15) and acute lymphoblastic leukemia (ALL, n = 3) at the time of
232
diagnosis. Interestingly, while ALL cases were nearly negative for ESAM, 10 of 15 AML
233
cases were clearly ESAM-positive (Figure 6B). Notably, the ESAM expression pattern was
234
completely different in the same AML category (Figure 6B). Thus, AML cells may change
235
their ESAM expression levels according to the cell-intrinsic features and/or the surrounding
236
environment.
AC C
237
EP
231
238
KG-1a leukemic cells express ESAM concomitant with the acquisition of a more
239
aggressive phenotype
240
The discrepancy between AML cell lines and primary AML cells with respect to
11
ACCEPTED MANUSCRIPT ESAM expression indicates that this antigen is differentially regulated in vivo. Therefore, we
242
inoculated ESAM– KG-1a cell lines into NOD/SCID mice and harvested reconstituted KG-1a
243
(rKG-1a) cells 10 weeks after inoculation. The cells were then cultured in vitro for 3 weeks
244
and inoculated again into NOD/SCID mice (Figure 7A). These rKG-1a cells also showed
245
high leukemic activity (Figure 7B). Flow cytometry analyses revealed that although parental
246
KG-1a cells were ESAM-negative, rKG-1a cells showed high ESAM expression (Figure 7C).
247
To investigate the significance of ESAM up-regulation in rKG-1a cells, ESAM– and ESAM+
248
rKG-1a cells were purified and inoculated into sub-lethally irradiated NOD/SCID mice.
249
Three months after transplantation, all mice were sacrificed, and the BM cells were analyzed.
250
Flow cytometry analyses revealed that ESAM– rKG-1a cells did not engraft, whereas ESAM+
251
rKG-1a cells engrafted (Figure 7D and 7E). These results indicate that leukemia cells
252
fluctuate in their surface phenotype according to surrounding environment and that ESAM
253
expression is related to the affinity of leukemia cells to BM stromal cells.
SC
M AN U
TE D EP AC C
254
RI PT
241
12
ACCEPTED MANUSCRIPT 255 256
Discussion Precisely defining the immunophenotype of HSCs is critical for the successful outcome of transplantation therapy for hematological malignancies. In addition, recent
258
progress in regenerative therapy using induced pluripotent stem cells demands that reliable
259
cell surface antigens are available to distinguish authentic HSCs from lineage-specific
260
progenitors [20, 21]. However, it is more difficult to characterize human HSCs and their
261
proximate progenitors compared with murine HSCs, in part because of the limited
262
information available regarding human HSC surface antigens [2, 22]. The developmental
263
process of HSCs with respect to ontogeny and transcription factors is thought to be similar
264
between mice and humans. Therefore, identifying common HSC antigens shared by the two
265
species will greatly facilitate basic and clinical studies of HSC biology. In this study, we
266
found that ESAM is a robust and cross-species HSC marker. Furthermore, the molecule is
267
functional on HSCs and is associated with specific subsets of leukemia cells.
TE D
M AN U
SC
RI PT
257
Using strict phenotypic definitions of human HSCs and their proximate progenitors
269
in the CD34+ CD38– fraction [14], we found that the CD90+ CD45RA– HSCs expressed high
270
levels of ESAM, and that this expression decreased during their differentiation into lymphoid
271
progenitors. In our culture and transplantation experiments, we also showed that high ESAM
272
expression segregates with multi-lineage potential and SCID-repopulating HSCs in humans;
273
these data are consistent with our previous findings in mice [11]. In addition, and similar to
274
the case for murine HSCs, ESAM was evidently a robust marker of human HSCs regardless
275
of changes to their functional properties and expression of other cell surface antigens. We
276
also assessed the long-term reconstitution capacity of ESAMHigh CD34+ CD38– cells from
277
various tissues (BM, GMPB, and CB) simultaneously and observed comparable results (data
278
not shown).
AC C
EP
268
13
ACCEPTED MANUSCRIPT Our previous study highlighted the functional importance of ESAM in the
280
proliferation and differentiation of HSCs. ESAM expression is markedly increased on murine
281
HSCs following cytotoxic insult [12]. Additionally, ESAM-deficient mice are less tolerant to
282
5-FU treatment, and the recovery of their HSC fraction was significantly delayed compared
283
with wild-type mice [12]. Furthermore, we found that ESAM is vital for the early stages of
284
erythrocyte recovery after BM injury (T. Sudo, manuscript in preparation). To date, few
285
functional markers that are commonly expressed on both human and murine HSC have been
286
reported. Thus, ESAM is important not only as a marker but also for determining the identity
287
of HSCs across species.
SC
M AN U
288
RI PT
279
Recently CD166, a member of the immunoglobulin superfamily, was reported as a universal marker for murine and human HSCs [23]. This molecule was also expressed on
290
endosteal niche components and immature osteoblasts, which exhibit high HSC-supporting
291
activity [24, 25]. Reciprocal transplantation of CD166-deficient HSCs into wild-type mice
292
and wild-type HSCs into CD166-deficient mice indicated that hemophilic CD166-CD166
293
interaction likely mediates the HSC-niche association and affects HSC maintenance. Given
294
that ESAM is also a member of the immunoglobulin superfamily with hemophilic interacting
295
features, it may also be involved in such HSC-niche interactions. However, because ESAM is
296
an endothelial lineage-related antigen, this molecule may mediate the interaction between
297
HSCs and endothelial cells. Indeed, irradiation or 5-FU treatment up-regulates ESAM
298
expression on murine HSCs, resulting in an intimate association between ESAMHigh HSCs
299
and the vasculature of BM [12]. In this context, both irradiation and 5-FU decreased CD166
300
expression on HSCs [23]. Therefore, we propose that these two markers are important for
301
HSC maintenance but they do not necessarily have overlapping functions.
302
AC C
EP
TE D
289
Although it is widely accepted that human HSCs reside in the CD34+ CD38–
14
ACCEPTED MANUSCRIPT fraction, recent studies reported that a CD34+ CD38Low fraction also contains human
304
hematopoietic repopulating cells [26, 27]. Our flow cytometry analysis revealed high ESAM
305
expression on CD34+ CD38Low cells (Figure 1A, right). Thus, high ESAM expression may
306
enrich HSCs regardless of CD38 expression. Furthermore, long-term reconstitution activity
307
has also been observed in the CD34– population [28-31]. Previous studies by Sonoda et al.
308
have verified that cells negative for 18 lineage-related antigens and positive for CD133
309
effectively enrich SCID-repopulating HSC activity in human CB CD34– populations [32-34].
310
Since ESAM expression covers a broader range of CB cells that are negative for
311
lineage-related antigens compared to CD34 expression (T. Yokota, unpublished observation),
312
ESAM as well as CD133 may be useful as positive markers for the CD34– HSCs. Future
313
studies will provide a more accurate marker set for estimating HSC numbers in human CB.
SC
M AN U
314
RI PT
303
Human CB contains various types of progenitor cells that are useful for tissue regenerative therapy [35]. For example, the CB CD34+ fraction can contribute to vasculature
316
development in zebrafish [36]. Furthermore, the administration of human CB CD34+ cells
317
ameliorates neurological damage after brain injury in rodents, presumably by stimulating
318
angiogenesis [37]. Thus, CB CD34+ cells likely contain a substantial amount of angiopoietic
319
progenitors. In the present study, we identified a new subpopulation of CB CD34+ CD38–
320
cells that express high levels of ESAM; these cells are apparently related to the endothelial
321
lineage. The ‘ESAMBright’ phenotype may be useful for enriching angiopoietic progenitors.
322
However, thorough examination of their differentiation potential in vitro and in vivo is
323
required to evaluate this possibility.
324
AC C
EP
TE D
315
Since high ESAM expression was related to the totipotent nature and robust growth
325
potential of normal HSCs, we expected that ESAM would be expressed in many leukemia
326
cell lines. However, all tested myeloid and lymphoid cell lines were ESAM-negative. In
15
ACCEPTED MANUSCRIPT 327
contrast, erythroid and megakaryocyte cell lines exhibited robust ESAM expression. Given
328
that ESAM is indispensable for normal erythropoiesis and thrombocyte function [12, 38], the
329
molecule may be intrinsically involved in the erythrocyte and megakaryocyte lineages. We observed that the human AML cell line KG-1a, which is normally ESAM–,
RI PT
330
became ESAM+ concomitant with engraftment in immunodeficient mice. Although we
332
cannot exclude the possibility that serial xenogeneic transplantation induced some
333
transformation in KG-1a cells, there are other biological explanations for this phenotypic
334
change. For example, ESAM may be expressed in a rare subpopulation of KG-1a cells that
335
comprise the leukemic stem cell (LSC) compartment. In this context, a recent approach for
336
identifying LSC markers revealed ESAM in the plasma membrane proteome of
337
LSC-enriched fractions obtained from myeloid leukemia patients with poor prognosis [39].
338
Furthermore, such rare ESAM+ KG-1a cells may acquire higher proliferative activity in vivo
339
by taking advantage of the interaction with the leukemia-supporting microenvironment in the
340
BM [40]. Indeed, ESAM+ rKG-1a cells showed higher engraftment capacity compared with
341
ESAM– rKG-1a cells. In this case, targeting the leukemia-microenvironment interaction via
342
ESAM may be a therapeutic strategy, as ESAM may confer a more aggressive clinical course
343
in some types of leukemia. As shown in Figure 6, the pattern of ESAM expression varied
344
significantly among primary AML cases. Although we observed no correlation between
345
ESAM expression and clinical features at this stage, further studies and longer observation
346
periods may reveal the true clinical value of ESAM in the diagnosis and treatment of acute
347
leukemia.
348
AC C
EP
TE D
M AN U
SC
331
In summary, we demonstrated that ESAM is a robust functional marker of HSCs in
349
humans as well as in mice. Additionally, ESAM is expressed on some types of human
350
leukemia cells and may be useful for lineage determination and as a prognostic indicator. The
16
ACCEPTED MANUSCRIPT 351
cross-species properties of ESAM expression will accelerate the translation of findings in
352
animal models to studies in humans; this will ultimately increase our understanding of the
353
identity and biology of human HSCs and leukemia cells.
RI PT
354 Acknowledgments
356
We thank Dr. Satoko Fujita (Obstetrics and Gynecology, Osaka University) and Dr. Takashi
357
Sakai (Orthopedic Surgery, Osaka University) for providing human samples. This work was
358
supported by Japan Society for the Promotion of Science (JSPS) KAKENHI (Grant Numbers
359
24591423 and 26461446).
M AN U
SC
355
360 Author contributions
362
T.I. and T.Y. designed and performed the research, analyzed the data, and wrote the
363
manuscript; Y.K. designed the research, analyzed the data, and wrote the manuscript; H.T.
364
established rKG-1a cells; M.I., T.S., and Y.S. designed and performed the microarray
365
analyses, and analyzed the data. Y.D., T.U., A.T., Y.H., S.E., H.S., and K.O. designed the
366
research and analyzed the data.
EP
367
TE D
361
Conflicts of interest disclosure
369
The authors declare no conflict of interest.
370 371
AC C
368
372
Supplementary methods
373
Mononuclear cell (MNC) preparation
374
Cord blood was diluted 1:1 with Dulbecco’s phosphate-buffered saline without
17
ACCEPTED MANUSCRIPT calcium and magnesium (PBS(–); Nacalai Tesque, Kyoto, Japan) and was overlaid onto an
376
equivalent volume of Ficoll-Paque PLUS (GE Healthcare Life Sciences, Little Chalfont, UK).
377
Cells were centrifuged for 25 min at 20°C at 400 ×g. MNCs were isolated and washed twice
378
with RPMI 1640 medium (Nacalai Tesque).
RI PT
375
379 380
Flow cytometry analysis and cell sorting
Allophycocyanin (APC)-conjugated anti-CD34 (HPCA-2), fluorescein
382
isothiocyanate (FITC)-conjugated anti-CD38 (HIT2) and CD45 (HI30), and Phycoerythrin
383
(PE)-conjugated anti-CD4 (SK3), CD8 (SK1), CD13 (WM15), and CD33 (P67-6)
384
monoclonal antibodies (mAbs) were purchased from BD Biosciences (San Diego, CA, USA).
385
PE-conjugated anti-CD45 (HI30), CD90/Thy1 (5E10), CD133 (AC133), Flk1/VEGFR-2
386
(7D4-6), VE-cadherin (BV9), and CD19 (HIB19) were purchased from BioLegend (San
387
Diego, CA, USA). ESAM mAb (Clone 408519) was purchased from R&D Systems
388
(Minneapolis, MN, USA) and biotinylated using EZ-Link Sulfo-NHS-Biotin and
389
biotinylation kits (Thermo Scientific, Waltham, MA, USA). Streptavidin-conjugated with
390
PE-Texas Red (Life Technologies Corp., Carlsbad, CA, USA), PE, and APC (BD
391
Biosciences) were used for visualization of biotinylated antibodies. Dead cells were excluded
392
by staining with 7-amino actinomycin D (7-AAD; Calbiochem, San Diego, CA, USA). Cells
393
were washed and then resuspended in PBS(–) containing 3% fetal bovine serum (MP
394
Biomedicals, Irvine, CA, USA). Cells were analyzed using FACSAria or FACSCanto II (BD
395
Biosciences, Franklin Lakes, NJ, USA). In some experiments, CD34+ CD38– ESAM–/Low,
396
CD34+ CD38– ESAMHigh, and CD34+ CD38– ESAMBright cells were sorted using a FACSAria
397
and used in subsequent experiments. The FACS data was analyzed using FlowJo software
398
(Tree Star, San Carlos, CA, USA).
AC C
EP
TE D
M AN U
SC
381
18
ACCEPTED MANUSCRIPT 399 400
Colony-forming cell assays
401
For myeloid colony formation, cells were cultured for 14 days in MethoCult H4434 (StemCell Technologies, Vancouver, Canada). Subsequently, colonies were enumerated and
403
classified as colony-forming unit-granulocyte-macrophage (CFU-GM), burst-forming
404
unit-erythroid (BFU-E), or CFU-Mix according to their shape and color, as observed under
405
an inverted microscope. All cultures were incubated at 37°C in a humidified chamber with
406
5% CO2.
SC
RI PT
402
408 409
M AN U
407 Limiting dilution assays
Cord blood CD34+ CD38– ESAMHigh or CD34+ CD38– ESAMBright cells were plated at various concentrations from 1 to 100 cells/well. Each well contained 200 µL MSC growth
411
medium with 10 ng/mL stem cell factor (SCF) and 5 ng/mL Flt-3 ligand. Half of the culture
412
medium was replaced with fresh medium containing the same cytokines twice per week.
413
After 28 days of culture, wells containing cells that had expanded were scored.
416
EP
415
SCID-repopulation assays
NOD/Shi-scid, IL-2γnull (NOG) mice were purchased from the Central Institute for
AC C
414
TE D
410
417
Experimental Animals (Kawasaki, Kanagawa, Japan). Sorted CB cells (CD34+ CD38–
418
ESAM–/Low, ESAMHigh, or ESAMBright) were injected via the tail vein after whole-body
419
irradiation at a dose of 1.8 Gy. After 3 months, BM cells were obtained and analyzed by flow
420
cytometry. Whole BM MNCs (2 × 106 cells/mouse) from primary recipients were injected
421
into secondary recipients via the tail vein after whole-body irradiation. After 4 months, BM
422
cells were obtained and analyzed by flow cytometry. Animal studies were performed with the
19
ACCEPTED MANUSCRIPT 423
approval of the Institutional Review Board of Osaka University.
424
426
Microarray Total RNAs were extracted from CD34+ CD38– ESAMHigh or ESAMBright cells of
RI PT
425
collagenase-treated BM using an RNeasy Micro Kit (Qiagen, Hilden, Germany). RNA
428
samples were subjected to gene expression analysis using a SurePrint G3 Human GE 8 × 60K
429
v2 Microarray (Agilent Technologies, Santa Clara, CA, USA).
SC
427
430
432
KG-1a mouse leukemia model
M AN U
431
KG-1a cells derived from a human acute myeloid leukemia (AML) sample were transplanted into the tail vein of 2 Gy-irradiated NOD/SCID mice. Ten weeks after
434
transplantation, reconstituted KG-1a (rKG-1a) cells were obtained from the BM of recipients.
435
The rKG-1a cells were cultured in vitro for 3 weeks, and then transplanted into the tail vein
436
of 2 Gy-irradiated NOD/SCID mice.
EP
438
AC C
437
TE D
433
20
ACCEPTED MANUSCRIPT 439
References
440
1
441
mouse hematopoietic stem cells. Science. 1988;241:58-62. doi:10.1126/science.2898810
442
2
443
Hematopoietic Stem Cells: Histories and Recent Achievements. In: Pelayo R, ed. Advances
444
in Hematopoietic Stem Cell Research. Rijeka, Croatia: InTech; 2012. p. 77-88.
445
doi:10.5772/32381
446
3
447
family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial
448
niches for stem cells. Cell. 2005;121:1109-1121. doi:10.1016/j.cell.2005.05.026
449
4
450
and enigma. Stem Cells. 2007;25:1339-1347. doi:10.1634/stemcells.2006-0644
451
5
452
hematopoietic stem cells cannot be purified based only on SLAM family markers. Blood.
453
2011;117:1550-1554. doi:10.1182/blood-2009-03-212803
454
6
455
generations of hematopoietic colonies derived from single nonlineage-committed
456
CD34+CD38- progenitor cells. Blood. 1991;77:1218-1227.
457
7
458
retention identifies long-term human hematopoietic stem cells within the Lin-CD34+CD38-
459
population. Blood. 2007;109:543-545. doi:10.1182/blood-2006-06-030270
460
8
461
defines a population of long-term reconstituting hematopoietic stem cells. Blood.
Spangrude GJ, Heimfeld S, Weissman IL. Purification and characterization of
SC
RI PT
Yokota T, Oritani K, Butz S, Ewers S, Vestweber D, Kanakura Y. Markers for
M AN U
Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM
Holmes C, Stanford WL. Concise review: stem cell antigen-1: expression, function,
TE D
Larochelle A, Savona M, Wiggins M, et al. Human and rhesus macaque
AC C
EP
Terstappen LW, Huang S, Safford M, Lansdorp PM, Loken MR. Sequential
McKenzie JL, Takenaka K, Gan OI, Doedens M, Dick JE. Low rhodamine 123
Randall TD, Lund FE, Howard MC, Weissman IL. Expression of murine CD38
21
ACCEPTED MANUSCRIPT 462
1996;87:4057-4067.
463
9
464
hematopoietic stem cells changes during developmental stages. Blood. 2001;97:419-425.
465
doi:10.1182/blood.V97.2.419
466
10
467
and CD34 by adult murine hematopoietic stem cells. Blood. 2001;97:2618-2624.
468
doi:10.1182/blood.V97.9.2618
469
11
470
hematopoietic progenitors throughout life in mice. Blood. 2009;113:2914-2923.
471
doi:10.1182/blood-2008-07-167106
472
12
473
hematopoietic stem cell status between quiescence and self-renewal. J Immunol.
474
2012;189:200-210. doi:10.4049/jimmunol.1200056
475
13
476
hematopoietic stem cell marker. Stem Cells. 2009;27:653-661.
477
doi:10.1634/stemcells.2008-0824
478
14
479
the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early
480
lymphoid development. Nat Immunol. 2010;11:585-593. doi:10.1038/ni.1889
481
15
482
hematopoietic stem and progenitor cells. Blood. 1997;90:5002-5012.
483
16
484
stem cell antigen: isolation, characterization, and molecular cloning. Blood.
485
1997;90:5013-5021.
RI PT
Matsuoka S, Ebihara Y, Xu M, et al. CD34 expression on long-term repopulating
SC
Tajima F, Deguchi T, Laver JH, Zeng H, Ogawa M. Reciprocal expression of CD38
M AN U
Yokota T, Oritani K, Butz S, et al. The endothelial antigen ESAM marks primitive
TE D
Sudo T, Yokota T, Oritani K, et al. The endothelial antigen ESAM monitors
EP
Ooi AG, Karsunky H, Majeti R, et al. The adhesion molecule esam1 is a novel
AC C
Doulatov S, Notta F, Eppert K, Nguyen LT, Ohashi PS, Dick JE. Revised map of
Yin AH, Miraglia S, Zanjani ED, et al. AC133, a novel marker for human
Miraglia S, Godfrey W, Yin AH, et al. A novel five-transmembrane hematopoietic
22
ACCEPTED MANUSCRIPT 486
17
Choi KD, Vodyanik MA, Togarrati PP, et al. Identification of the hemogenic
487
endothelial progenitor and its direct precursor in human pluripotent stem cell differentiation
488
cultures. Cell Rep. 2012;2:553-567. doi:10.1016/j.celrep.2012.08.002
489
18
490
co-stimulatory and adhesion molecules and chemokine or apoptosis receptors on acute
491
myeloid leukaemia: high CD40 and CD11a expression correlates with poor prognosis. Br J
492
Haematol. 2001;115:298-308. doi:10.1046/j.1365-2141.2001.03085.x
493
19
494
risk classification in AML independent of established biomarkers: ECOG phase 3 trial,
495
E1900. Blood. 2012;120:2297-2306. doi:10.1182/blood-2012-02-414425
496
20
497
pluripotent stem cells. Int J Hematol. 2012;95:617-623. doi:10.1007/s12185-012-1094-x
498
21
499
advances and challenges toward de novo generation of hematopoietic stem cells. Blood.
500
2013;122:4035-4046. doi:10.1182/blood-2013-07-474825
501
22
502
Curr Opin Hematol. 2013;20:265-272. doi:10.1097/MOH.0b013e3283612628
503
23
504
hematopoietic stem cells and the hematopoietic niche. Blood. 2014;124:519-529.
505
doi:10.1182/blood-2014-03-565721
506
24
507
niche cell populations that regulate hematopoietic stem cells. Blood. 2010;116:1422-1432.
508
doi:10.1182/blood-2009-08-239194
509
25
SC
RI PT
Brouwer RE, Hoefnagel J, Borger van Der Burg B, et al. Expression of
M AN U
Gonen M, Sun Z, Figueroa ME, et al. CD25 expression status improves prognostic
Togarrati PP, Suknuntha K. Generation of mature hematopoietic cells from human
TE D
Slukvin, II. Hematopoietic specification from human pluripotent stem cells: current
EP
Zhang Q, Iida R, Yokota T, Kincade PW. Early events in lymphopoiesis: an update.
AC C
Chitteti BR, Kobayashi M, Cheng Y, et al. CD166 regulates human and murine
Nakamura Y, Arai F, Iwasaki H, et al. Isolation and characterization of endosteal
Chitteti BR, Cheng YH, Kacena MA, Srour EF. Hierarchical organization of
23
ACCEPTED MANUSCRIPT 510
osteoblasts reveals the significant role of CD166 in hematopoietic stem cell maintenance and
511
function. Bone. 2013;54:58-67. doi:10.1016/j.bone.2013.01.038
512
26
513
highly variable proliferation and self-renewal properties comprise the human hematopoietic
514
stem cell compartment. Nat Immunol. 2006;7:1225-1233. doi:10.1038/ni1393
515
27
516
CD38 on CD34-positive human hematopoietic repopulating cells. Exp Hematol.
517
2007;35:1429-1436. doi:10.1016/j.exphem.2007.05.017
518
28
519
hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in
520
multiple species. Nat Med. 1997;3:1337-1345. doi:10.1038/nm1297-1337
521
29
522
human hematopoietic cells with SCID-repopulating activity. Nat Med. 1998;4:1038-1045.
523
doi:10.1038/2023
524
30
525
characterization of human CD34(-)Lin(-) and CD34(+)Lin(-) hematopoietic stem cells using
526
cell surface markers AC133 and CD7. Blood. 2000;95:2813-2820.
527
31
528
blood-derived CD34- cells assured by intra-bone marrow injection. Blood.
529
2003;101:2924-2931. doi:10.1182/blood-2002-09-2782
530
32
531
blood-derived CD34(-) SCID-repopulating cells using intra-bone marrow injection.
532
Leukemia. 2010;24:162-168. doi:10.1038/leu.2009.206
533
33
RI PT
McKenzie JL, Gan OI, Doedens M, Wang JC, Dick JE. Individual stem cells with
SC
McKenzie JL, Gan OI, Doedens M, Dick JE. Reversible cell surface expression of
M AN U
Goodell MA, Rosenzweig M, Kim H, et al. Dye efflux studies suggest that
TE D
Bhatia M, Bonnet D, Murdoch B, Gan OI, Dick JE. A newly discovered class of
EP
Gallacher L, Murdoch B, Wu DM, Karanu FN, Keeney M, Bhatia M. Isolation and
AC C
Wang J, Kimura T, Asada R, et al. SCID-repopulating cell activity of human cord
Kimura T, Matsuoka Y, Murakami M, et al. In vivo dynamics of human cord
Ishii M, Matsuoka Y, Sasaki Y, et al. Development of a high-resolution purification
24
ACCEPTED MANUSCRIPT 534
method for precise functional characterization of primitive human cord blood-derived
535
CD34-negative SCID-repopulating cells. Exp Hematol. 2011;39:203-213 e201.
536
doi:10.1016/j.exphem.2010.11.008
537
34
538
distinct class of primitive human cord blood-derived CD34-negative hematopoietic stem cells.
539
Leukemia. 2014;28:1308-1315. doi:10.1038/leu.2013.326
540
35
541
mesenchymal stem cells are enriched at different gestational ages in human umbilical cord
542
blood. Pediatr Res. 2008;64:68-73. doi:10.1203/PDR.0b013e31817445e9
543
36
544
human CD34+ progenitor cells in zebrafish. Arterioscler Thromb Vasc Biol.
545
2011;31:1589-1597. doi:10.1161/ATVBAHA.111.226969
546
37
547
umbilical cord blood CD34(+) cells in a mouse model of neonatal stroke. Neuroscience.
548
2014;263:148-158. doi:10.1016/j.neuroscience.2014.01.018
549
38
550
(ESAM) localizes to platelet-platelet contacts and regulates thrombus formation in vivo. J
551
Thromb Haemost. 2009;7:1886-1896. doi:10.1111/j.1538-7836.2009.03606.x
552
39
553
proteomics and transcriptomics approach to identify leukemic stem cell (LSC) markers. Mol
554
Cell Proteomics. 2013;12:626-637. doi:10.1074/mcp.M112.021931
555
40
556
Br J Haematol. 2014;164:767-778. doi:10.1111/bjh.12725
557
41
RI PT
Takahashi M, Matsuoka Y, Sumide K, et al. CD133 is a positive marker for a
M AN U
SC
Javed MJ, Mead LE, Prater D, et al. Endothelial colony forming cells and
Pozzoli O, Vella P, Iaffaldano G, et al. Endothelial fate and angiogenic properties of
TE D
Tsuji M, Taguchi A, Ohshima M, et al. Effects of intravenous administration of
AC C
EP
Stalker TJ, Wu J, Morgans A, et al. Endothelial cell specific adhesion molecule
Bonardi F, Fusetti F, Deelen P, van Gosliga D, Vellenga E, Schuringa JJ. A
Tabe Y, Konopleva M. Advances in understanding the leukaemia microenvironment.
Ciraci E, Della Bella S, Salvucci O, et al. Adult human circulating
25
ACCEPTED MANUSCRIPT 558
CD34(-)Lin(-)CD45(-)CD133(-) cells can differentiate into hematopoietic and endothelial
559
cells. Blood. 2011;118:2105-2115. doi:10.1182/blood-2010-10-316596
560 Figure legends
562
Figure 1. Expression of ESAM on human BM and GMPB.
563
Mononuclear cells (MNCs) from human BM were stained with anti-CD34, anti-CD38, and
564
anti-ESAM antibodies and analyzed by flow cytometry (A). The ESAM expression patterns
565
of the CD34+ CD38– fraction of BM (B) and GMPB (C) are shown. The solid black lines and
566
dashed lines show ESAM and background levels, respectively. The CD34+ CD38– fraction
567
was subdivided into two populations: ESAM–/Low and ESAMHigh, and the percentages of cells
568
in each gate are shown in each panel. (D) CD34+ CD38– cells in ABM were stained with
569
CD45RA and CD90 (Thy-1) and subdivided into 3 fractions, including CD45RA– CD90+
570
hematopoietic stem cells (HSC); CD45RA– CD90– multipotent progenitors; and CD45RA+
571
CD90– multi-lymphoid progenitors (MLP) (left panel). The percentage of cells in each
572
fraction is shown in the upper right section of the panel. In the middle panel, ESAM
573
expression levels in HSCs (solid line), multipotent progenitors (dashed line), and MLPs
574
(dotted line) are shown. Grey line shows background level of CD34+ CD38– cells.
575
Differences between background and mean ESAM intensities in each fraction are shown
576
(right panel).
SC
M AN U
TE D
EP
AC C
577
RI PT
561
578
Figure 2. High ESAM expression marks primitive hematopoietic progenitors.
579
(A) ESAM–/Low and ESAMHigh cells of the CD34+ CD38– fraction of BM were sorted and
580
subjected to a methylcellulose colony formation assay. The numbers of colony-forming units
26
ACCEPTED MANUSCRIPT 581
(CFUs) are shown. CFU-G/GM/M, CFU- granulocyte/granulocyte-macrophage/macrophage;
582
BFU-E, burst-forming unit erythrocyte; CFU-Mix, mixed colony-forming unit. (B) ESAM–
583
/Low
584
with MS5 in the presence of stem cell factor, Flt3 ligand, and interleukin-7. Cell numbers of
585
each fraction after 1–4 weeks are shown. *p < 0.05; **p < 0.005 by Student’s t-test. (C)
586
Numbers of CD19+ CD13– B-lineage cells after 4-week-co-culture with MS5 are shown.
587
****p < 0.0001 by Student’s t-test.
588
SC
RI PT
and ESAMHigh cells of the CD34+ CD38– fraction of BM were sorted and co-cultured
Figure 3. Expression of ESAM on human CB cells.
590
(A) CB MNCs were stained with anti-CD34 and CD38 antibodies and analyzed by flow
591
cytometry (left panel). The percentages of cells in each subpopulation are shown in the upper
592
right section of the panel. ESAM expression on CD34– CD38+, CD34+ CD38+, CD34– CD38–,
593
and CD34+ CD38– fractions were analyzed by flow cytometry (right panel). According to
594
ESAM expression levels, the CD34+ CD38– fraction was subdivided into three populations:
595
ESAM–/Low, ESAMHigh, and ESAMBright (lower right). The same gates were applied to other
596
fractions, and the percentages of cells in each gate are shown in each panel. The solid black
597
lines and dashed lines show ESAM and background levels, respectively. (B) Ten CB samples
598
were analyzed in the same manner, and the percentages of ESAM–/Low, ESAMHigh, and
599
ESAMBright cells are shown. The percentage of each population of the same CB sample is
600
represented by the same symbols.
TE D
EP
AC C
601
M AN U
589
602
Figure 4. High ESAM expression marks authentic HSCs in CB.
603
(A) ESAMHigh and ESAMBright cells in the CB CD34+ CD38– fraction were sorted and
604
subjected to methylcellulose colony formation assay. Numbers of colony-forming units
27
ACCEPTED MANUSCRIPT (CFUs) are shown. BFU-E, burst-forming unit erythrocyte; CFU-GM/M, colony-forming unit
606
granulocyte-macrophage/macrophage; CFU-Mix, mixed colony-forming unit. (B) ESAMHigh
607
and ESAMBright cells of CB CD34+ CD38– fraction were subjected to limiting dilution
608
analyses in the MS5 co-culture system in the presence of stem cell factor and
609
granulocyte-colony-stimulating factor (G-CSF). The input cell numbers corresponding to
610
37% negative value are shown in rectangles. (C) ESAM–/Low, ESAMHigh, and ESAMBright cells
611
in the CB CD34+ CD38– fraction were purified and transplanted into sub-lethally irradiated
612
NOG mice (ESAM–/Low 5 × 103 cells/mouse, ESAMHigh 2 × 104 cells/mouse, ESAMBright 1 ×
613
103 cells/mouse, n = 6 for each group). Three months after transplantation, all recipients were
614
sacrificed and flow cytometry analyses of BM were performed. BM MNCs were stained with
615
mouse CD45 and human CD45, and the percentages of human CD45+ cells are shown. (D)
616
The representative results of the recipient transplanted ESAMHigh cells are shown. Human
617
CD45+ cells were gated and the percentages of each lineage marker-positive cells are shown.
618
(E) Whole BM MNCs were obtained from primary recipients of ESAMHigh cells and were
619
transplanted into sub-lethally irradiated NOG mice (2 × 106 cells/mouse, n = 5). Four months
620
after transplantation, all recipients were sacrificed and flow cytometry analyses of BM were
621
performed. The percentages of human CD45+ cells are shown. (F) The figures shows
622
representative results for the secondary recipient.
SC
M AN U
TE D
EP
AC C
623
RI PT
605
624
Figure 5. Morphological, immunophenotypic, and gene expression profiling differences
625
between ESAMHigh and ESAMBright cells of the CD34+ CD38– fraction.
626
(A) ESAM–/Low, ESAMHigh and ESAMBright cells of CB CD34+ CD38– fraction were sorted,
627
and cytospin slides were stained by May-Grünwald/Giemsa staining. Horizontal bars
628
represent 20 µm. (B) CB CD34+ CD38– cells were stained with ESAM and CD45,
28
ACCEPTED MANUSCRIPT hematopoietic cell marker (left panel), and CD133, HSC marker (right panel). (C) ESAMHigh
630
and ESAMBright cells of the CB CD34+ CD38– fraction were back-plotted onto the CD34 and
631
CD38 profile. ESAMHigh cells were found as CD34Low, whereas ESAMBright cells were
632
CD34High. (D) CB CD34+ CD38– cells were stained with ESAM and the endothelial markers,
633
VEGFR-2 (left panel) and VE-cadherin (right panel). (E) ESAM expression of the CD34+
634
CD38– fraction from collagenase-treated bone was determined by flow cytometry. ESAM–/Low,
635
ESAMHigh, and ESAMBright gates and the percentages of cells in each gate are shown. The
636
solid black line and dashed line show ESAM and background levels, respectively. (F) The
637
gene expression profiles of CD34+ CD38– ESAMHigh and ESAMBright cells from
638
collagenase-treated bone were determined by microarray. Heat maps of selected genes related
639
to endothelial and hematopoietic cells are shown. (G) The CD34+ CD38– ESAMBright cells
640
from collagenase-treated bone were cultured with MS5 stromal cells in the presence of VEGF,
641
stromal cell-derived factor (SDF) and IL-16. This co-culture condition supports both
642
endothelial and hematopoietic cell growth simultaneously [41]. After 5 weeks, the cells were
643
harvested and analyzed by flow cytometry. Recovered cells were stained with anti-CD34 and
644
CD45 antibodies (left panel). The expression of endothelial marker CD31 on the CD34+
645
CD45– cells is shown in the right panel.
646
AC C
EP
TE D
M AN U
SC
RI PT
629
647
Figure 6. ESAM expression on various human leukemia cell lines and primary leukemia
648
cells.
649
(A) The ESAM along with CD34 and CD38 expression patterns on various human leukemia
650
cell lines were analyzed by flow cytometry. (B) Primary leukemia cells were analyzed by
651
flow cytometry. Leukemia cells were gated in the CD34 and CD38 profiles, and the ESAM
652
expression patterns of leukemia cells are shown. The solid black lines and dashed lines show
29
ACCEPTED MANUSCRIPT 653
ESAM and background levels, respectively.
654 Figure 7. KG-1a leukemic mouse model.
656
(A) Scheme of the transplantation protocols. KG-1a, a human AML cell line, was
657
transplanted into sub-lethally irradiated NOD/SCID mice. Ten weeks after transplantation,
658
the mice were killed and leukemic cells in BM were collected. The reconstituted KG-1a
659
(rKG-1a) was cultured in vitro for 3 weeks and re-transplanted into sub-lethally irradiated
660
NOD/SCID mice. (B) Kaplan-Meier survival curves of transplanted mice are shown. (C)
661
ESAM expression pattern of KG-1a and rKG-1a were analyzed by flow cytometry by using a
662
monoclonal antibody specific for human ESAM. The solid black line and dashed line show
663
ESAM expression level in rKG-1a and KG-1a, respectively. (D) ESAM– and ESAM+ rKG-1a
664
cells were sorted and transplanted into sub-lethally irradiated NOD/SCID mice (5 × 104
665
cells/mouse, n = 4 for each group). Three months after transplantation, the mice were
666
sacrificed and BM cells were analyzed by flow cytometry. The percentages of human CD45+
667
cells are shown. *p < 0.05 by Student’s t-test. (E) Representative results of the mice
668
transplanted with ESAM– and ESAM+ rKG-1a cells are shown. The percentages of human
669
CD45+ cells (mean ± SD) are shown in the rectangles.
SC
M AN U
TE D
EP
AC C
670
RI PT
655
30
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
EP
Figure 1. CD34+ cells
BM MNCs
A 10 5
10 5
21
10 3
2
0
3
10
2
0
10 2
0
10 3
10 4
10 5
CD34
B
C
80
% of Max
80
60
40
40
16.7
83.3
20
0 0
10
2
10
3
10
4
10
5
0
ESAM
10
2
10
3
10
4
10
5
ESAM 300
100
105
HSC
24
MPP
80 104
60
13 % of Max
HSC 103
102
MLP 60
40
200
100
20
MLP 103
CD45RA
104
105
0 0 102
103
ESAM
104
105
0
MLP
0
MPP
MPP
HSC
CD90
10 5
60
89.6
0
0
10 4
mobilized PB 100
20
D
10 3
CD38
100
10.4
10 2
0
adult BM
% of Max
10
⊿ESAM Mean
10
AC C
10 4
ESAM
ESAM
10 4
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ESAMHigh
30
20
6
4
10
2
0
0
-E
H ig h
w
ES
AM
Lo –/
AM
1
2
3 Week
4
1000 0 H ig h
* 0
0
2000
w
1 x105
3000
AM
**
****
4000
Lo
2 x105
1x104
5000
–/
**
C
ES
ESAM –/Low ESAMHigh
*
AM
4 x105
*
3 x105
ES
Cell Number
2x104
ES
B
CD19+ CD13– Cell Number
C
FU
-G
/G
BF
U
M
Colonies / 200 input cells
8
ESAM–/Low
/M
Colonies / 200 input cells
40
M ix
A
AC C
Figure 2.
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
Figure 3.
CD34–CD38+
A
10
4
34 0.1 10
3
10
2
80
80
60
CD38
60
69
40 10
10
1
0
10
0
1
10
10
2
10
3
10
CD34+CD38+
100
100
66 0.2
4
20
20
0
0 0
4
10
10
1
10
2
10
3
10
4
% of Max
100
100
80
80
60
10
1
10
2
10
23
10
3
10
4
60
91
40
9
40
8
0.5
33
59
20
0
0 0
10
1
10
2
10
3
ESAM 100
% of CD34+ CD38– cells
0
68
CD34+CD38–
20
80
60
40
20
0
ESAM –/Low
10
CD34–CD38–
CD34
B
7
40
27
ESAM High
ESAM Bright
4
10
0
10
10
1
10
2
10
3
10
4
Figure 4. CFU-Mix BFU-E CFU-GM/M
120
12
0
20
100
37
100
10
80
% negative
Colony counts / 1000 cells
Input cell number
B
140
60
40
40
60
80
100
ESAM Bright
1
20
63
ESAM High
EP
A
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ESAM High ESAM Bright
D
BM MNC
10
Mouse CD45
40
20
0
10
4
10
3
43
B lymphoid
300
200
200
15
72
15
50 100
100
10 2
10 3
10 4
10 5
0
0 0
10
2
10
3
10
4
10
5
0
10
2
10
3
10
4
10
5
0
10
2
10
3
10
4
10
5
rig ht
ig h
300
0
ES AM B
/L
ES AM H
400
100
0
ES AM –
T lymphoid
400 150
10 2
0
E
Human CD45
F
Human CD13 and CD33
Human CD19
Human CD4 and CD8
BM MNC
15
Myeloid
10 5
B lymphoid
T lymphoid
12 12
10 4
5
0
15
1.1
9 9
# Cells
Mouse CD45
10
10 3
10
16
6 6
ig h
12
77
5
10 2
3
3
0 0
ES AM H
human CD45 % of BM MNCs
Myeloid
5
# Cells
60
ow
human CD45 % of BM MNCs
C
AC C
0.1
0
0
10 2
10 3
Human CD45
10 4
10 5
0 0
10
2
10
3
10
4
10
5
Human CD13 and CD33
0 0
10 2
10 3
Human CD19
10 4
10 5
0
10 2
10 3
10 4
Human CD4 and CD8
10 5
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
E
ESAMBright cells
100 80 60 40
4
CD133
2
10
2
3
0 10
4
10
10
5
10
10
2
3
0 10
4
10
ESAMHigh 10
4
10
4
10
3
3
10
2
0 2
0 10
3
10
4
10
2
10
0
5
2
10
3
0 10
CD34
10
4
10
5
10
4
10
VEGFR2
3
10
2
10
0 2
0 10
3
10
ESAM
4
10
5
10
ht rig
MH
MB A
ES A
HSC self-renewal and expansion
Hematopoietic
5
2
G
5
10
HSC development
10
CD34
10
KDR HHEX FLI1 TEK PECAM1 ESAM FLT1 EMCN TIE1 LMO2 TAL1 CBFB RUNX1 PBX1 EZH2 TCEA1 PTEN MEIS1 SOX17 CITED2 GATA2 MYB GFI1 BMI1 CBFA2T3 HOXB4 HBE1 HBG1 GYPA ITGA2B NFE2 VAV1 GATA1 MPO
Angiohematopoietic
3
4
5
100
5
10
4
10
3
10
80
4
% of Max
CD38
10 10
105
ig h
ht
Typical endothelial genes
5
104
rig
10
ESAMBright
5
103
A MB
5
10
ESAM
10
CD38
h
0
ESAM
VE-cadherin
ig
2
10
3
10
CD45
CD45
10
F11R JAM3 TJP1 TJP2 CDH5 CDH2 CLDN5 HIF1A AAMP F2R EDF1 PROCR SCARF1 NOS3 CAV1 S1PR1 CTGF APOLD1 PPAP2B AMOTL1 AMOTL2 AMOT TNFAIP1 COL18A1 MMRN1 SERPINE1 MALL ANGPT1 ANGPTL2 ROBO4 EPAS1 PGF ENG
Endothelial junctions
3
0
D
A MH
4
10
3
0 102
ESAM
ES
10
10
C
F
5
10
0
ES
5
AC C
B
60
20
20
20
ES
ESAMHigh cells
EP
ESAM–/Lowcells
% of Max
A
TE D
Figure 5.
2
10
0
60 40
2
10
20
0 2
0 10
3
10
ESAM
4
10
5
10
2
0 10
3
10
CD34
4
10
5
10
0
2
0 10
3
10
CD31
4
10
5
10
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
Figure 6. A
Myeloid 10 4
10 3
10
10 2
10 2
0
10 4
10 5
10 5
10 5
10 4
10 4
10 3
10 3
10 2
10 2
10 3
10 4
10 5
10 4
10 4
3
10
10 2
2
10
3
10
4
10
5
10
10 2
10 3
10 4
10 5
0
10 2
10 3
10 4
10 5
10 5
10 5
10 4
10 4
10 3
10 3
10 2
10 5
10
2
10
0
3
10
4
10
5
2
10
3
10
4
10
5
Erythroid / Megakaryocyte
K562
3
0
10
10
2
10
3
10
4
10
5
2
0
0
10 2
10 3
10 4
10 5
10 2
0
10 5
10 5
10 4
10 4
10
3
10 3
10
2
10
0
2
0
0
10 2
10 3
10 4
10 5
10 5
10 5
10 4
10 4
10 3
10 3
10 4
10 5
10 3
10 2
10 2
0 10 5
0
10 2
10 3
10 4
10 5
0
10 2
10 3
10 4
10 5
0
10 2
10 3
10 4
10 5
0
10 5 0
4
10
CMK 3
10
10 2
0
0
10
2
10
3
10
4
10
5
10 3
10 4
10 5
10 5
10 5
10 4
10
4
10 3
10
3
2
10
2
3
10 2
0
10 2
4
0
10
2
10
3
10
4
10
ESAM
ESAM
10
10 4
10 3
10 2 0
5
10
0
0
CD34
CD38 0
10
2
10
3
10
4
10
5
CD34
B
0
10 3
10 2
0
HEL
10 3
10
10 2
10 4
10 4
10
0
10 5
10 5
Kasumi
3
10 2
0
10 2
0
10
10 4
3
0
10 2
0
ESAM
10 5
MOLT4
0
10 5
0
KG1a
10 4
AC C
ESAM
10 2
10 5
10
10 3
0
0
10
0
10 2
0
ESAM
10 3
0
THP1
3
EP
ESAM
U937
10 2
10 5
10 4
0
0
10 5
TE D
10 4
Lymphoid Jurkat ESAM
10 5
ESAM
10 5
ESAM
ESAM
HL60
AML (M2)
AML (M2)
APL
CD38
APL
ALL
% of Max
100 80 60 40 20 0 0 10
2
10
3
ESAM
10
4
10
5
0 10
2
10
3
ESAM
10
4
10
5
0 10
2
10
3
ESAM
10
4
10
5
0 10
2
10
3
ESAM
10
4
10
5
0 10
2
10
3
ESAM
10
4
10
5
KG-1a
1.0×106
80
cells
i.v. 10 weeks
60 40 20
0 0 10
bone marrow from leukemic mouse
10
3
10
4
10
5
ESAM
E in vitro culture for 3 weeks
Mouse CD45
reconstituted KG-1a (rKG-1a) 5.0×105 cells i.v.
B
ESAM– rKG-1a 10 5
10 4
10 4
10
3
10
KG-1a (n=5) rKG-1a cl 1 (n=5)
60
rKG-1a cl 2 (n=5)
40 20 0 0
2
4
6
8
Weeks after transplantation
10
0.1
0.0
10 2
3
10 2
0.0056 ± 0.00324
0
10
2
10
3
10
4
10
5
0.1634 ± 0.0586
0
0
10
Human CD45
80
0.2
ESAM+ rKG-1a
10 5
0
100
% Survival
2
*
0.3
ES AM –
NOD/SCID
D
100
ES AM +
C
2.4 Gy
% of Max
A
Human CD45 % of BM MNCs
Figure 7.
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
2
10
3
10
4
10
5
ACCEPTED MANUSCRIPT
ESAM marks hematopoietic stem cells in humans as well as in mice.
RI PT
ESAM enriches human hematopoietic stem cells in all clinically available sources.
AC C
EP
TE D
M AN U
SC
ESAM marks a subset of human leukemia cells and reflects some of their features.