ArticlePDF Available

Human Hematopoietic Stem Cells Can Survive In Vitro for Several Months

Wiley
Advances in Hematology
Authors:

Abstract and Figures

We previously reported that long-lasting in vitro hematopoiesis could be achieved using the cells differentiated from primate embryonic stem (ES) cells. Thus, we speculated that hematopoietic stem cells differentiated from ES cells could sustain long-lasting in vitro hematopoiesis. To test this hypothesis, we investigated whether human hematopoietic stem cells could similarly sustain long-lasting in vitro hematopoiesis in the same culture system. Although the results varied between experiments, presumably due to differences in the quality of each hematopoietic stem cell sample, long-lasting in vitro hematopoiesis was observed to last up to nine months. Furthermore, an in vivo analysis in which cultured cells were transplanted into immunodeficient mice indicated that even after several months of culture, hematopoietic stem cells were still present in the cultured cells. To the best of our knowledge, this is the first report to show that human hematopoietic stem cells can survive in vitro for several months.
Content may be subject to copyright.
Hindawi Publishing Corporation
Advances in Hematology
Volume 2009, Article ID 936761, 7pages
doi:10.1155/2009/936761
Research Article
Human Hematopoietic Stem Cells Can
Survive In Vitro for Several Months
Tar o I s hi g ak i ,1, 2 Kazuhiro Sudo,1Takashi Hiroyama,1Kenichi Miharada,1
Haruhiko Ninomiya,2Shigeru Chiba,2Toshiro Nagasawa,2and Yukio Nakamura1
1Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074, Japan
2Division of Hematology, Institute of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
Correspondence should be addressed to Yukio Nakamura, yukionak@brc.riken.jp
Received 22 September 2008; Revised 8 December 2008; Accepted 15 December 2008
Recommended by N. Chao
We previously reported that long-lasting in vitro hematopoiesis could be achieved using the cells dierentiated from primate
embryonic stem (ES) cells. Thus, we speculated that hematopoietic stem cells dierentiated from ES cells could sustain long-lasting
in vitro hematopoiesis. To test this hypothesis, we investigated whether human hematopoietic stem cells could similarly sustain
long-lasting in vitro hematopoiesis in the same culture system. Although the results varied between experiments, presumably due
to dierences in the quality of each hematopoietic stem cell sample, long-lasting in vitro hematopoiesis was observed to last up to
nine months. Furthermore, an in vivo analysis in which cultured cells were transplanted into immunodeficient mice indicated that
even after several months of culture, hematopoietic stem cells were still present in the cultured cells. To the best of our knowledge,
this is the first report to show that human hematopoietic stem cells can survive in vitro for several months.
Copyright © 2009 Taro Ishigaki et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
Identification of in vitro culture protocols that enable
somatic stem cells to survive and proliferate will be of value
not only for basic research but also clinical applications that
require somatic stem cells. The development of an ecient
method for in vitro proliferation of mesenchymal stem cells,
for example, has allowed cultured mesenchymal stem cells to
be used in clinical applications [1].
Although hematopoietic stem cells have been extensively
analyzed and characterized [2], in vitro proliferation of these
cells remains problematic using established culture methods
[3,4]. In addition, the length of time that hematopoietic
stem cells can survive in an in vitro culture system remains
uncertain. CD34-positive (CD34+) cells have been identified
in long-term in vitro cultures of hematopoietic stem cells [5
9]. However, as none of these previous studies performed an
in vivo assay of the cultured cells, such as transplantation into
mice, it is uncertain whether hematopoietic stem cells with
the capacity to reconstitute long-term in vivo hematopoiesis
were present in these prolonged in vitro cultures.
We previously described a culture method that pro-
duced long-lasting in vitro hematopoiesis using non-human
primate embryonic stem (ES) cells [10]. We speculated
that hematopoietic stem cells derived from ES cells could
sustain long-lasting in vitro hematopoiesis. To test this
hypothesis, we initiated long-term in vitro cultures of human
hematopoietic stem cells using the same culture method
as previously [10]. In addition, we evaluated the in vivo
function of cells cultured in vitro for several months by
transplanting them into immunodeficient mice.
2. Materials and Methods
2.1. Cell Culture. We purchased human umbilical cord blood
samples from the Cell Engineering Division of RIKEN
BioResource Center (Tsukuba, Ibaraki, Japan). The ethical
committee of the RIKEN Tsukuba Institute approved the
use of human umbilical cord blood before the study was
initiated. CD34+hematopoietic stem/progenitor cells were
collected from human umbilical cord blood using a magnetic
cell sorting system, MACS CD34 Isolation kit (Miltenyi
2Advances in Hematology
Biotec Inc., Sunnyvale, Calif, USA), according to the man-
ufacturer’s instructions.
Mouse-derived cell lines (OP9 and C3H10T1/2) were
purchased from the Cell Engineering Division of RIKEN
BioResource Center (Tsukuba, Ibaraki, Japan) and were
cultured as follows: OP9 in Minimum Essential Medium-
α(MEM-α; Invitrogen, Carlsbad, Calif, USA) containing
20% fetal bovine serum (FBS; Invitrogen, Calif, USA);
C3H10T1/2 in Basal Medium Eagle (BME; Invitrogen)
containing 10% FBS (BioWest, Miami, Fla, USA). The cell
lines were γ-irradiated (50 Gy) before use as feeder cells.
CD34+cells were cultured on feeder cells in a 100 mm
dish in Iscove’s modified Dulbeccos medium (IMDM;
SIGMA, St Louis, Mo, USA) containing 10% FBS (BioW-
est), 10 μg/mL bovine insulin, 5.5 μg/mL human transfer-
rin, 5 ng/mL sodium selenite (ITS liquid MEDIA supple-
ment; SIGMA-Aldrich, Mass, USA), 100unit/mL penicillin,
100 μg/mL streptomycin, 2 mM L-glutamine (PSQ; Invit-
rogen), 50 ng/mL stem cell factor (SCF; R&D Systems,
Minneapolis, Minn, USA), 50 ng/mL Flt-3 ligand (Flt-
3L; R&D Systems), and 50 ng/mL thrombopoietin (TPO;
R&D Systems). The initial number of CD34+cells placed
in culture varied between experiments: 5 ×103cells in
Exp-OP9-A, Exp-10T1/2-A, and Exp-OP9-B; 4 ×104cells
in Exp-OP9-F and Exp-10T1/2-F; 5 ×104cells in Exp-
OP9-C, Exp-OP9-E, Exp-10T1/2-E, Exp-OP9-H, and Exp-
10T1/2-H; 8 ×104cells in Exp-OP9-G and Exp-10T1/2-
G; 1 ×105cells in Exp-10T1/2-I; 2 ×105cells in Exp-
OP9-D and Exp-10T1/2-D. The letters A to “I” after
Exp-OP9 or Exp-10T1/2 indicate 9 dierent umbilical cord
blood samples derived from 9 dierent neonates. Samples
A, B, C, F, G, H, and I were frozen after collection,
while samples D and E were used immediately as fresh
samples.
Twenty-four hours after initiation of culture, the
medium together with any detached cells was removed
and fresh medium was added to the culture. Thereafter,
the medium was changed every 3-4 days (twice a week).
The number of cells attached to the feeder cells increased
gradually. Approximately four weeks after initiation of
culture, attached cells were dissociated using a 0.25% trypsin
EDTA solution (SIGMA-Aldrich) and cultured again on new
feeder cells. Thereafter, similar passages of attached cells were
performed every 3-4 weeks.
The number of viable cells was assessed using an
automated cell counter and an assay based on the trypan
blue dye exclusion method, ViCell (BECKMAN COULTER,
Fullerton, Calif, USA). The morphology of the cells was
analyzed by microscopic examination after Wright staining
(Muto Pure Chemicals, Tokyo, Japan).
2.2. Flow Cytometry. Cells were stained with monoclonal
antibodies (MoAbs) and analyzed using a FACS Calibur
(BD Biosciences, San Jose, Calif, USA). The following
MoAbs were purchased from BD Biosciences: fluores-
cein isothiocyanate- (FITC-) conjugated MoAb against
human CD14 (FITC-CD14), FITC-CD34, FITC-CD41a,
and FITC-CD45; phycoerythrin-conjugated MoAb against
human CD4(PE-CD4), PE-CD11b, PE-CD13, PE-CD34,
PE-CD45, PE-CD56, and PE-CD235a (Glycophorin A);
allophycocyanin-conjugated MoAb against human CD3
(APC-CD3), APC-CD8, APC-CD19, APC-CD33, and APC-
CD45. PE-CD33 was purchased from eBiosciences (San
Diego, Calif, USA). FITC-mouse IgG1, PE-mouse IgG1,
APC-mouse IgG1, FITC-mouse IgG2a, and PE-mouse IgG2b
were also purchased from BD Biosciences and were used as
isotype controls. Cell viability was monitored by staining
with propidium iodide (SIGMA-Aldrich). Flow cytometry
data were analyzed using CellQuest (BD Biosciences) analysis
software.
2.3. Colony Formation Assay. Cells (1 ×104)werecultured
in a 35 mm dish with 1 mL of Methocult (H4435; Stem cell
technology, Vancouver, British Columbia, Canada) for 10–14
days, and the number of separate colonies was determined
by macroscopic morphology. Representative colonies were
picked up and cell morphology was analyzed by microscopic
examination after Wright staining (Muto Pure Chemicals).
2.4. Cell Transplantation into Mice. Eight-week-old male
NOD/Shi-scid IL-2Rγnull (NOG) mice were purchased from
the Central Institute for Experimental Animals (Kawasaki,
Kanagawa, Japan) and used within two weeks of delivery
in all experiments. Prior to cell transplantation, the mice
were given a sublethal dose of γ-rays (3.0 Gy). A 200 μL
cell suspension in phosphate-buered saline (PBS; SIGMA)
containing 5% FBS (BioWest) was injected intravenously
into the tail vein of each mouse. All experimental procedures
on the mice were approved by the Institutional Animal Care
and Use Committee of the RIKEN Tsukuba Institute.
3. Results and Discussion
3.1. Long-Lasting In Vitro Hematopoiesis from Human
Hematopoietic Stem Cells. Human CD34+cells were cultured
on feeder cells in the presence of SCF, Flt-3L, and TPO. We
used the mouse-derived cell lines, OP9 and C3H10T1/2, as
feeder cells. Both OP9 [1015] and C3H10T1/2 [10,16
18] have been used in many studies to maintain in vitro
hematopoiesis. As we show below, both OP9 and C3H10T1/2
cells supported long-lasting in vitro hematopoiesis from
human hematopoietic stem cells.
About one week after initiation of culture, cobblestone
areas were observed on the feeder cells (Figures 1(a)
and 1(b)), indicating that human hematopoietic cells were
proliferating. The numbers of cells attached to the feeder
cells increased gradually (Figures 1(c) and 1(d)) and the
numbers of detached cells, derived from the attached cells,
also increased gradually. When the medium was changed,
the numbers of detached cells in the medium were counted
and the cells were subjected to analyses such as flow
cytometric analysis. Detached cells were removed during the
medium changes and were not cultured further in any of the
experiments.
Detached cells were continuously produced for several
months in experiments Exp-OP9-A, Exp-OP9-F, Exp-OP9-
H, Exp-10T1/2-A, and Exp-10T1/2-H (Figure 2). As we
Advances in Hematology 3
(a) (b) (c) (d)
Figure 1: Appearance of the cells attached to feeder cells. Representative examples of CD34(+) human hematopoietic stem/progenitor cells
cultured on either OP9 feeder cells ((a), (c)) or C3H10T1/2 feeder cells ((b), (d)) for 7 days ((a), (b)) or 22 days ((c), (d)).
0 70 140 210 280
Days of culture
Exp-OP9-A
0 70 140 210 280
Days of culture
Exp-OP9-B
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-OP9-C
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-OP9-D
0.001
0.01
0.1
1
10
×103
0.001
0.01
0.1
1
10
×103
#
Numbers of detached cells produced from CD34(+)cells
0 70 140 210 280
Days of culture
Exp-OP9-E
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-OP9-F
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-OP9-G
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-OP9-H
0.001
0.01
0.1
1
10
×103
Numbers of detached cells produced from CD34(+)cells
(a)
0 70 140 210 280
Days of culture
Exp-10T1/2-A
0 70 140 210 280
Days of culture
Exp-10T1/2-D
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-10T1/2-E
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-10T1/2-F
0.001
0.01
0.1
1
10
×103
0.001
0.01
0.1
1
10
×103
#
Numbers of detached cells produced from CD34(+)cells
0 70 140 210 280
Days of culture
Exp-10T1/2-G
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-10T1/2-H
0.001
0.01
0.1
1
10
×103
0 70 140 210 280
Days of culture
Exp-10T1/2-I
0.001
0.01
0.1
1
10
×103
Numbers of detached cells produced from CD34(+)cells
(b)
Figure 2: Production of hematopoietic cells in long-term cultures of human hematopoietic stem cells. (a) Eight independent experiments
were performed using eight dierent umbilical cord blood samples and OP9 cells as feeder cells. (b) Seven independent experiments were
performed using seven dierent umbilical cord blood samples and C3H10T1/2 cells as feeder cells. ((a), (b)) The number of detached cells
in the overlying medium was counted at each medium change (approximately half weekly). The data are shown as the mean number of
detached cells produced from a single CD34(+) cell, that is, the total number of detached cells divided by the number of CD34(+) cells used
to initiate the culture. Exp: experiment. A to I after Exp-OP9 and Exp-10T1/2 indicate 9 dierent umbilical cord blood samples derived from
9dierent neonates. #: Cultures Exp-OP9-A and Exp-10T1/2-F were terminated because of fungal infection.
detail below, flow cytometric analysis and a transplantation
assay demonstrated that the detached cells produced in
this culture method included both mature and imma-
ture hematopoietic cells, such as colony-forming cells and
hematopoietic stem cells. We found that production of
detached cells eventually ceased in all experiments except
for Exp-OP9-A. Unfortunately, we were forced to halt Exp-
OP9-A because of fungal infection although the cells in this
culture proliferated eciently and robustly before fungal
contamination (Figure 2).
The numbers of detached cells varied among the exper-
iments (Figure 2) and, notably, showed no correlation with
the initial number of CD34+cells used in each experiment.
Thus, 5×103CD34+cellswereusedinExp-OP9-AandExp-
10T1/2-A and both cultures produced substantial numbers
of detached cells over a prolonged period (Figure 2). In
contrast, a larger number of CD34+cells (2 ×105)wasused
to initiate the Exp-OP9-D and Exp-10T1/2-D cultures, but
they produced considerably fewer detached cells (Figure 2).
These results indicate that the rate of production of detached
4Advances in Hematology
CD33
CD34
On OP9 On C3H10T1/2
CD33
CD34
(a)
Day 57
0
20
40
Numberof colonies
Day 60
0
20
40
Numberof colonies
Day 127
0
20
40
Numberof colonies
On OP9
Day 134
0
20
40
Numberof colonies
On C3H10T1/2
(b)
Figure 3: Characterization of cultured cells. (a) Flow cytometric analysis of detached cells produced in cultures on OP9 (Exp-OP9-A) and
C3H10T1/2 (Exp-10T1/2-A) feeder cells and collected on Day 218 of culture. The detached cells were stained for CD33, a marker specific
for granulocyte/macrophage lineage cells, and CD34, a marker specific for hematopoietic stem/progenitor cells. Flow cytometric analyses
of detached cells from other experiments showed similar results. (b) Colony-formation assays. Detached cells produced in culture on OP9
feeder cells (Exp-OP9-A) were collected on Days 57 and 127 of culture. Similarly, detached cells produced in culture on C3H10T1/2 feeder
cells (Exp-10T1/2-D) were collected on Days 60 and 134 of culture. The cell samples were used in a standard colony-formation assay. Black
bars: colony-forming unit of monocyte/macrophage lineage cells, CFU-M. Blue bars: colony-forming unit of granulocyte lineage cells, CFU-
G. Yellow bars: colony-forming unit of granulocyte and monocyte/macrophage lineage cells, CFU-GM. Red bars: burst-forming unit of
erythroid cells, BFU-E. Similar results were obtained in colony-formation assays using detached cells from other cultures.
cells depended on the quality rather than the number of
CD34+cells used in each experiment. In other words, when
the quality of CD34+cells was high, 5 ×103CD34+cells
were sucient to generate ecient in vitro hematopoiesis as
shown in Exp-OP9-A and Exp-10T1/2-A (Figure 2).
The majority of detached cells had the morphological
characteristics of granulocyte/macrophage lineage cells (Sup-
plementary Figure S1, available at doi 10.1155/2009/936761)
although some blast-like cells were also present. Con-
sistent with their morphological phenotype, the major-
ity of the detached cells expressed CD33, a marker
of granulocyte/macrophage lineage cells (Figure 3(a)). Of
note, CD34+CD33+cells, which are less mature than
CD34CD33+cells, were abundant among the detached cells
even at 7 months after initiation of culture (Figure 3(a)).
A colony-formation assay demonstrated that granulo-
cyte, macrophage, and erythrocyte progenitor cells were
present among the detached cells (Figure 3(b)). As men-
tioned above, when the culture medium was changed,
detached cells were removed and were not cultured further.
Instead, they were either used in experimental analyses or
discarded. As is shown in Figure 2, detached cells were
continuously produced, and they included abundant colony-
forming cells even at Day 127 and Day 134. As one
example, the calculated total number of colony-forming
cells present in detached cells at Day 127 (upper right,
Figure 3(b)) was 13 311, which corresponded to a greater
than ten-fold increase in the numbers of colony-forming cells
compared to the starting material of this culture, that is,
983 colony-forming cells in 5 ×103CD34+cells. Thus, the
culture method we describe here could continuously produce
abundant colony-forming cells for several months.
Although a mixed colony (a colony derived from
very immature hematopoietic cells) was not observed in
the colony formation assay (Figure 3(b)), it nevertheless
remained possible that hematopoietic stem cells were present
at a very low frequency among the detached cells. Therefore,
we performed a transplantation assay in which detached cells
were injected into an immunodeficient NOD/Shi-scid IL-
2Rγnull (NOG) mouse (mentioned hereafter).
3.2. Hematopoietic Stem Cells Cultured In Vitro for Several
Months Give Rise to Long-Lasting In Vivo Hematopoiesis
after Transplantation into Mice. Detachedcellswerecollected
from Exp-OP9-A on Day 169 after initiation of culture
and transplanted into an NOG mouse (3.9×106cells)
(Figure 4(a)). Peripheral blood samples from the mouse
were subjected to flow cytometric analysis on Days 56 and
Advances in Hematology 5
CD33
CD34(+) cells (Exp-OP9-A)
In vitro culture for 169 days
Detached cells produced for 3.5 days
Transplantation into mouse
In vivo hematopoiesis for 126 days
Sacrificed and analyzed
CD13
(a)
CD45
CD33
(b)
CD13
CD33
(c)
CD13
CD33
CD14
CD11b
CD45
CD19
(d)
CD34(+) cells (Exp-OP9-F)
In vitro culture for 125 days
Detached cells produced for 3.5 days
Transplantation into mouse
In vivo hematopoiesis for 184 days
Sacrificed and analyzed
CD34
CD33
(e)
CD33
CD19
Gly-A
CD3
CD14
CD11b
CD4
CD56
CD41a
CD8
(f)
Figure 4: Flow cytometric analysis of hematopoietic cells of the mouse that had been transplanted with human hematopoietic cells
produced by in vitro culture. ((a), (e)) Schema of the experimental procedure and flow cytometric analysis of transplanted cells. ((a)–
(d)) Detached cells (3.9×106cells) produced on OP9 feeder cells (Exp-OP9-A) were collected on Day 169 of culture and transplanted
into an immunodeficient NOG mouse. Peripheral blood was collected on Days 56 (b) and 112 (c) after transplantation, and bone marrow
cells were collected on Day 126 after transplantation (d). ((e), (f)) Detached cells (2.4×106cells) produced on OP9 feeder cells (Exp-
OP9-F) were collected on day 125 of culture and transplanted into an immunodeficient NOG mouse. The bone marrow cells were
collected on Day 184 after transplantation and were analyzed. ((a)–(f)) The cells were stained using monoclonal antibodies against CD45, a
leukocyte common antigen, CD34, a marker specific for hematopoietic stem/progenitor cells, CD33 and CD13, markers of granulocyte and
monocyte/macrophage lineage cells, CD11b and CD14, markers of monocyte/macrophage lineage cells, CD19, a marker of B lymphocyte
lineage cells, CD3, CD4, and CD8, markers of T lymphocyte lineage cells, Gly-A (Glycophorin A), a marker of erythroid cells, CD56, a
marker of large granular lymphocytes and natural killer cells, and CD41a, a marker of megakaryocyte/platelet lineage cells.
6Advances in Hematology
112 after transplantation. Human hematopoietic cells were
clearly present in the peripheral bloods (Figures 4(b), 4(c)).
The mouse was sacrificed on Day 126 after transplantation,
and bone marrow and spleen cells were subjected to flow
cytometric analysis. Human hematopoietic cells were present
in the bone marrow (Figure 4(d)) but were present at a
very low rate in spleen (data not shown). The estimated
rate of chimerism of human CD45+cells in bone marrow
was 2.6% when compared to the number of mouse CD45+
cells. The human hematopoietic cells detected in the bone
marrow included cells of the myeloid lineage (CD13+CD33+:
9.7% of the human CD45+cells), the monocyte/macrophage
lineage (CD11b+CD14+: 3.8% of the human CD45+cells),
the B cell lineage (CD19+: 80.5% of the human CD45+cells)
(Figure 4(d)), and other lineages at very low levels (data not
shown).
Detached cells were collected from Exp-OP9-F on
day 125 after initiation of culture and transplanted into
an NOG mouse (2.4×106cells) (Figure 4(e)). CD45+
human hematopoietic cells were present in peripheral blood
from the mouse one month after transplantation (data
not shown). The mouse was sacrificed on Day 184 after
transplantation, and the bone marrow cells were subjected
to flow cytometric analysis. The rate of chimerism of human
CD45+cells in the bone marrow was 0.2% when compared
to the number of mouse CD45+cells.Thebonemarrow
contained human hematopoietic cells, which included cells
of the myeloid lineage (CD33+: 23.5% of the human CD45+
cells), the monocyte/macrophage lineage (CD11b+CD14+:
6.7% of the human CD45+cells), as well as the B (CD19+:
53.7% of the human CD45+cells) and T cell lineages
(CD3+,CD4
+,CD8
+, and CD4+CD8+: 10.7% of the human
CD45+cells) (Figure 4(f)). Erythroid (Glycophorin A+)and
megakaryocyte (CD41a+)lineagecellswerepresentatvery
low rates (Figure 4(f)).
In the transplantation assay described above, human
hematopoietic cells of various lineages were present in mice
up to six months after cell transplantation. In general,
it is impossible that in vivo hematopoiesis derived from
transplanted cells is maintained for several months solely by
committed progenitor cells. Therefore, the transplanted cells
appeared to include hematopoietic stem cells, that is, even
after in vitro culture for several months, hematopoietic stem
cells were still present in cultures Exp-OP9-A and Exp-OP9-
F. In light of the numbers of cells produced in culture and of
the duration of this production, the hematopoietic stem cells
also appeared to have survived for several months in Exp-
OP9-H, Exp-10T1/2-A, and Exp-10T1/2-H.
The in vitro expansion of human hematopoietic stem
cells is known to be very dicult [3]. In agreement with
this, a mixed colony (a colony derived from very imma-
ture hematopoietic cells) was not observed in the colony
formation assay in this study (Figure 3(b)), indicating that
hematopoietic stem cells did not expand to any great extent
in our culture method. However, since it was highly unlikely
that in vitro hematopoiesis could be maintained for several
months solely by committed progenitor cells present in the
starting materials, the long-lasting in vitro hematopoiesis
was likely maintained by hematopoietic stem cells.
On the basis of a previous estimation of the numbers
of hematopoietic stem cells capable of repopulating in
NOD/SCID mice [19], the starting materials we used in
Exp-OP9-A and Exp-OP9-F should have included a very
low number of NOD/SCID-repopulating cells. However,
the transplantation assay demonstrated that NOD/SCID-
repopulating cells were present among the detached cells
that were continuously produced in our culture method
(Figure 4), strongly suggesting that our culture method
continuously produced small numbers of new NOD/SCID-
repopulating cells throughout the long-term culture period.
Hence, the total number of NOD/SCID-repopulating cells
that were produced as detached cells throughout the whole
long-term in vitro culture was likely greater than the number
of such cells that were present in the starting materials.
Taken together, the hematopoietic stem cells capable
of repopulating in NOD/SCID mice in our culture system
appeared to be maintained by asymmetric cell division, that
is, one of the daughter cells retained the characteristics of
hematopoietic stem cells and another did not.
Leukemic stem cells might also survive and/or proliferate
in our culture method for a prolonged period, enabling basic
research or screening for eective anticancer drugs to be
performed on these cultured cells. In addition, basic research
on specific diseases, such as aplastic anemia or paroxysmal
nocturnal hemoglobinuria, might benefit from a long-term
culture system for hematopoietic stem cells derived from
patients.
4. Conclusions
To the best of our knowledge, this is the first report to show
that human hematopoietic stem cells can survive in vitro for
several months. Since the duration of in vitro hematopoiesis
appeared to depend on the quality of hematopoietic stem
cells present in each sample, our culture method may be of
value for assessing the quality of hematopoietic stem cells
prior to their use in the clinic. In particular, our method
could be used for the evaluation of umbilical cord bloods
since these samples are routinely used in the clinic following
preservation for several months. For example, in this study
the quality of hematopoietic stem cells derived from samples
A, F, and H appeared to be higher compared to other
samples.
Acknowledgments
The authors obtained human umbilical cord blood from the
Cell Engineering Division of RIKEN BioResource Center,
which was supported by the National Bio-Resources Project
and the Stem Cell Resource Network (Banks at Miyagi,
Tokyo, Kanagawa, Aichi, and Hyogo) of the Ministry of
Education, Culture, Sports, Science, and Technology in Japan
(MEXT), and from Dr. Isamu Ishiwata (Ishiwata Hospital,
Mito, Ibaraki, Japan). This work was supported by grants
from MEXT. The authors thank all members in the Cell
Engineering Division for help, discussion, and secretarial
assistance.
Advances in Hematology 7
References
[1] K. Le Blanc, F. Frassoni, L. Ball, et al., “Mesenchymal stem cells
for treatment of steroid-resistant, severe, acute graft-versus-
host disease: a phase II study, The Lancet, vol. 371, no. 9624,
pp. 1579–1586, 2008.
[2] L. I. Zon, “Intrinsic and extrinsic control of haematopoietic
stem-cell self-renewal, Nature, vol. 453, no. 7193, pp. 306–
313, 2008.
[3] T. Heike and T. Nakahata, “Ex vivo expansion of hematopoi-
etic stem cells by cytokines, Biochimica et Biophysica Acta, vol.
1592, no. 3, pp. 313–321, 2002.
[4] K. Ando, T. Yahata, T. Sato, et al., “Direct evidence for ex
vivo expansion of human hematopoietic stem cells, Blood,
vol. 107, no. 8, pp. 3371–3377, 2006.
[5] T. M. Dexter, T. D. Allen, and L. G. Lajtha, “Conditions
controlling the proliferation of haemopoietic stem cells in
vitro, Journal of Cellular Physiology, vol. 91, no. 3, pp. 335–
344, 1977.
[6] I. D. Lewis, G. Almeida-Porada, J. Du, et al., “Umbilical cord
blood cells capable of engrafting in primary, secondary, and
tertiary xenogeneic hosts are preserved after ex vivo culture in
a noncontact system, Blood, vol. 97, no. 11, pp. 3441–3449,
2001.
[7]C.L.daSilva,R.Gonc¸alves,K.B.Crapnell,J.M.S.
Cabral,E.D.Zanjani,andG.Almeida-Porada,“Ahuman
stromal-based serum-free culture system supports the ex
vivo expansion/maintenance of bone marrow and cord blood
hematopoietic stem/progenitor cells, Experimental Hematol-
ogy, vol. 33, no. 7, pp. 828–835, 2005.
[8] P. Feugier, N. Li, D.-Y. Jo, et al., “Osteopetrotic mouse stroma
with thrombopoietin, c-kit ligand, and flk-2 ligand supports
long-term mobilized CD34+hematopoiesis in vitro, Stem
Cells and Development, vol. 14, no. 5, pp. 505–516, 2005.
[9] T. Yoshikubo, T. Inoue, M. Noguchi, and H. Okabe, “Dier-
entiation and maintenance of mast cells from CD34+human
cord blood cells, Experimental Hematology,vol.34,no.3,pp.
320–329, 2006.
[10] T. Hiroyama, K. Miharada, N. Aoki, et al., “Long-lasting in
vitro hematopoiesis derived from primate embryonic stem
cells,” Experimental Hematology, vol. 34, no. 6, pp. 760–769,
2006.
[11] T. Nakano, H. Kodama, and T. Honjo, “Generation of
lymphohematopoietic cells from embryonic stem cells in
culture, Science, vol. 265, no. 5175, pp. 1098–1101, 1994.
[12] T. Nakano, H. Kodama, and T. Honjo, “In vitro development
of primitive and definitive erythrocytes from dierent precur-
sors, Science, vol. 272, no. 5262, pp. 722–724, 1996.
[13] M.A.Vodyanik,J.A.Bork,J.A.Thomson,andI.I.Slukvin,
“Human embryonic stem cell-derived CD34+cells: ecient
production in the coculture with OP9 stromal cells and
analysis of lymphohematopoietic potential, Blood, vol. 105,
no. 2, pp. 617–626, 2005.
[14] T. Hiroyama, K. Miharada, K. Sudo, I. Danjo, N. Aoki,
and Y. Nakamura, “Establishment of mouse embryonic stem
cell-derived erythroid progenitor cell lines able to produce
functional red blood cells, PLoS ONE,vol.3,no.2,article
e1544, pp. 1–11, 2008.
[15] N. Takayama, H. Nishikii, J. Usui, et al., “Generation of
functional platelets from human embryonic stem cells in
vitro via ES-sacs, VEGF-promoted structures that concentrate
hematopoietic progenitors, Blood, vol. 111, no. 11, pp. 5298–
5306, 2008.
[16] G. Hangoc, R. Daub, R. G. Maze, J. H. Falkenburg, H. E. Brox-
meyer, and M. A. Harrington, “Regulation of myelopoiesis by
murine fibroblastic and adipogenic cell lines, Experimental
Hematology, vol. 21, no. 4, pp. 502–507, 1993.
[17] M. Nishikawa, K. Ozawa, A. Tojo, et al., “Changes in
hematopoiesis-supporting ability of C3H10T1/2 mouse
embryo fibroblasts during dierentiation, Blood, vol. 81, no.
5, pp. 1184–1192, 1993.
[18] T. L. Maekawa, T. A. Takahashi, M. Fujihara, et al., A novel
gene (drad-1) expressed in hematopoiesis-supporting stromal
cell lines, ST2, PA6 and A54 preadipocytes: use of mRNA
dierential display, Stem Cells, vol. 15, no. 5, pp. 334–339,
1997.
[19] T. Ueda, K. Tsuji, H. Yoshino, et al., “Expansion of human
NOD/SCID-repopulating cells by stem cell factor, Flk2/Flt3
ligand, thrombopoietin, IL-6, and soluble IL-6 receptor, The
Journal of Clinical Investigation, vol. 105, no. 7, pp. 1013–1021,
2000.
... Co-culture with feeder cells has also been shown to improve the hematopoietic output of hiPSCs [118]. Both OP9 and C3H10T1/2 feeders have seen use as supportive cells for differentiation [119][120][121]. Finally, the use of small molecules to enhance HSPC generation could be a potential solution to expensive cytokine cocktails. ...
Article
Full-text available
Amidst the global shortfalls in blood supply, storage limitations of donor blood and the availability of potential blood substitutes for transfusion applications, society has pivoted towards in vitro generation of red blood cells (RBCs) as a means to solve these issues. Many conventional research studies over the past few decades have found success in differentiating hematopoietic stem and progenitor cells (HSPCs) from cord blood, adult bone marrow and peripheral blood sources. More recently, techniques that involve immortalization of erythroblast sources have also gained traction in tackling this problem. However, the RBCs generated from human induced pluripotent stem cells (hiPSCs) still remain as the most favorable solution due to many of its added advantages. In this review, we focus on the breakthroughs for high-density cultures of hiPSC-derived RBCs, and highlight the major challenges and prospective solutions throughout the whole process of erythropoiesis for hiPSC-derived RBCs. Furthermore, we elaborate on the recent advances and techniques used to achieve cost-effective, high-density cultures of GMP-compliant RBCs, and on their relevant novel applications after downstream processing and purification.
... The most common feeder cells currently used are mouse embryonic fibroblasts (MEF) cells. Other cell types utilized for stem cell culture support are testicular stromal cells, as JK1, and embryonic mesenchymal stem cells (10T1/2) [8,9]. Alternatively, human adult uterine endometrial cells (hUECs), human adult breast parenchymal cells (hBPCs), and human embryonic fibroblasts (hEFs) have also been used as feeder cells [10,11]. ...
Article
Full-text available
Introduction We here present our findings on 2 types of feeder layers, one composed of mouse embryonic fibroblasts (MEF) and the second one of mouse skeletal myoblasts (C 2 Cl 2 ) feeder cells. Methods The 2 feeder layers present a dramatic variance of intrinsic stiffness (142.68 ± 17.21 KPa and 45.78 ± 9.81 KPa, respectively). Results and Conclusion This information could be used for a better understanding of cells and cell microenvironment mechano-physical characteristics that are influencing stem cell commitment, in order to develop a suitable engineered tissue for cardiac and skeletal muscle repair and a bio-actuator.
... HSCs are primarily located in the bone marrow and can be mobilized into the bloodstream which makes bone marrow and peripheral blood the common tissue sources for HSC extraction [62, 63••, 64]. Moreover, it has been shown that these cells can also be isolated from umbilical cord blood [65,66]. HSCs differentiate into the monocyte/macrophage lineage and further into osteoclasts under stimulation with receptor activator of nuclear factor kappa-B ligand (RANKL) and monocyte-colony-stimulating factors (M-CSF), both of which are secreted in vivo by osteoblasts and osteocytes [67,68]. ...
Article
Full-text available
Purpose of Review One aim in bone tissue engineering is to develop human cell-based, 3D in vitro bone models to study bone physiology and pathology. Due to the heterogeneity of cells among patients, patient’s own cells are needed to be obtained, ideally, from one single cell source. This review attempts to identify the appropriate cell sources for development of such models. Recent Findings Bone marrow and peripheral blood are considered as suitable sources for extraction of osteoblast/osteocyte and osteoclast progenitor cells. Recent studies on these cell sources have shown no significant differences between isolated progenitor cells. However, various parameters such as medium composition affect the cell’s proliferation and differentiation potential which could make the peripheral blood-derived stem cells superior to the ones from bone marrow. Summary Peripheral blood can be considered a suitable source for osteoblast/osteocyte and osteoclast progenitor cells, being less invasive for the patient. However, more investigations are needed focusing on extraction and differentiation of both cell types from the same donor sample of peripheral blood.
... It has been shown that OP9 feeder cells as a mouse bone marrow stromal cell line may enhance the hematopoietic differentiation of human iPSCs [86]. Also, C3H10T1/2 feeder cells have the capacity to stimulate the hematopoietic differentiation of human iPSCs [38,87]. Increasing in vitro evidence indicates that the cell type of origin and an epigenetic memory for iPSCs may influence on the hematopoietic differentiation of human iPSCs [34,88,89]. ...
Article
Full-text available
During the last years, several strategies have been made to obtain mature erythrocytes or red blood cells (RBC) from the bone marrow or umbilical cord blood (UCB). However, UCB-derived hematopoietic stem cells (HSC) are a limited source and in vitro large-scale expansion of RBC from HSC remains problematic. One promising alternative can be human pluripotent stem cells (PSCs) that provide an unlimited source of cells. Human PSCs, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are self-renewing progenitors that can be differentiated to lineages of ectoderm, mesoderm, and endoderm. Several previous studies have revealed that human ESCs can differentiate into functional oxygen-carrying erythrocytes; however, the ex vivo expansion of human ESC-derived RBC is subjected to ethical concerns. Human iPSCs can be a suitable therapeutic choice for the in vitro/ex vivo manufacture of RBCs. Reprogramming of human somatic cells through the ectopic expression of the transcription factors (OCT4, SOX2, KLF4, c-MYC, LIN28, and NANOG) has provided a new avenue for disease modeling and regenerative medicine. Various techniques have been developed to generate enucleated RBCs from human iPSCs. The in vitro production of human iPSC-derived RBCs can be an alternative treatment option for patients with blood disorders. In this review, we focused on the generation of human iPSC-derived erythrocytes to present an overview of the current status and applications of this field.
... The BM cells, BM-derived HSCs, and BM-MSCs were isolated from age-matched green fluorescent protein (GFP)-expressing transgenic mice with APPswe − /GFP + genetic background and cultured as described in previous reports [33][34][35]. The HSCs were cultured from the BM by utilizing an HSC enrichment set (BD Pharmingen) and cultured on OP9 cells as a feeder layer in Iscoves's modified Dulbecco's medium (IMDM) (Invitrogen) supplemented with 10 % fetal bovine serum (FBS), 20 ng/mL stem cell factor (Peprotech), 20 ng/ mL thrombopoietin (Peprotech), and 20 ng/mL Flt3-Ligand (Peprotech). ...
Article
Full-text available
Recent studies reported granulocyte colony-stimulating factor (G-CSF) treatment can improve the cognitive function of Alzheimer’s disease (AD) mice, and the mobilized hematopoietic stem cells (HSCs) or bone marrow mesenchymal stem cells (BM-MSCs) are proposed to be involved in this recovery effect. However, the exact role of mobilized HSC/BM-MSC in G-CSF-based therapeutic effects is still unknown. Here, we report that C-X-C chemokine receptor type 4 (CXCR4)/stromal cell-derived factor 1 (SDF-1) chemotaxis was a key mediator in G-CSF-based therapeutic effects, which was involved in the recruitment of repair-competent cells. Furthermore, we found both mobilized HSCs and BM-MSCs were able to infiltrate into the brain, but only BM-MSCs replenished the neural lineage cells and contributed to neurogenesis in the brains of AD mice. Together, our data show that mobilized BM-MSCs are involved in the replenishment of neural lineages following G-CSF treatment via CXCR4/SDF-1 chemotaxis and further support the potential use of BM-MSCs for further autogenically therapeutic applications.
... KMC iz popkovnične krvi lahko preživijo več mesecev in vitro, kar je ključnega pomena pri načrtovanju kontinuirane proizvodnje eritrocitov. 46 Erirociti, vzgojeni in vitro, razvijejo krvnoskupinske antigene v zaporedju, ki je ustrezno dogajanju in vivo, 47 imajo normalno izražene glikolitične encime, enako deformabilnost, enako preživetje v človeku in funkcionalen hemoglobin. 5,34,40 Izražanje HbF v eritrocitih in vitro bi lahko imelo pomen pri razvoju avtologne transfuzijske podpore pri hemoglobinopatijah, na primer pri anemiji srpastih celic, saj bi pridobili populacijo eritrocitov, ki bi v visokem odstotku izražali HbF, ki ne polimerizira tako kot HbS. 5 Imajo celo prednost pred alogenskimi eritrociti, saj so homogeni (sinhrono dozorevanje), njihova življenjska doba je blizu 120 dni, njihova predvidena razpolovna doba pa je daljša od povprečne razpolovne dobe alogenskih eritrocitov krvodajalca. ...
Article
Full-text available
Red cell transfusion is an established mode of therapy. Growing demand for red cell concentrates due to ageing population will in the next decades drive the introduction of new approaches in blood supply. In vitro red cell culturing is one of them. This technology enables us to yield red cells from various hematopoietic stem cells. The main goal is to gain large quantities of mature enucleated red cells expressing adult hemoglobin that are of »universal blood group« 0 RhD- negative and compatible with respect to the other blood group systems. At the moment, the technology efficiency does not allow the production of large amounts of red cells that could be comparable to blood donation gains.
... Stem cell factor interacts with specific c-kit receptor and triggers signalling cascade to promote haematopoiesis and stemness and also maintains suitable microenvironment of haematopoietic stem cells and progenitor cells. Basic fibroblast growth factor (bFGF) significantly reduces cellular senescence and promotes stem cell self-renewal and differentiation [58,[62][63][64][65][66][67][68]. The protocols for isolation and long-term proliferation of hUCBSCs have been well established now. ...
Article
Full-text available
The developing brain is found highly vulnerable towards the exposure of different environmental chemicals/drugs, even at concentrations, those are generally considered safe in mature brain. The brain development is a very complex phenomenon which involves several processes running in parallel such as cell proliferation, migration, differentiation, maturation and synaptogenesis. If any step of these cellular processes hampered due to exposure of any xenobiotic/drug, there is almost no chance of recovery which could finally result in a life-long disability. Therefore, the developmental neurotoxicity (DNT) assessment of newly discovered drugs/molecules is a very serious concern among the neurologists. Animal-based DNT models have their own limitations such as ethical concerns and lower sensitivity with less predictive values in humans. Furthermore, non-availability of human foetal brain tissues/cells makes job more difficult to understand about mechanisms involve in DNT in human beings. Although, the use of cell culture have been proven as a powerful tool for DNT assessment, but many in vitro models are currently utilizing genetically unstable cell lines. The interpretation of data generated using such terminally differentiated cells is hard to extrapolate with in vivo situations. However, human umbilical cord blood stem cells (hUCBSCs) have been proposed as an excellent tool for alternative DNT testing because neuronal development from undifferentiated state could exactly mimic the original pattern of neuronal development in foetus when hUCBSCs differentiated into neuronal cells. Additionally, less ethical concern, easy availability and high plasticity make them an attractive source for establishing in vitro model of DNT assessment. In this review, we are focusing towards recent advancements on hUCBSCs-based in vitro model to understand DNTs.
... The induction of hematopoietic differentiation on stromal cells [40] and formation of embryoid bodies (EB) [41,42] are the two experimental systems used to generate hematopoietic precursors from embryonic stem cells in most experiments [43]. We have previously modeled murine hematopoiesis using an embryonic stem cells (ES)/OP9 coculture which was shown to be a highly reproducible way to model hematopoiesis in vitro [40,44,45,46]. The OP9 stroma cell line provides the necessary extrinsic signals for the differentiation of pluripotent ES cells first into primitive flk1+ hemangioblasts (day 4–5) and then immature hematopoietic stem and progenitor cells (day 8). ...
Article
Full-text available
Somatic activating mutations in the Notch1 receptor result in the overexpression of activated Notch1, which can be tumorigenic. The goal of this study is to understand the molecular mechanisms underlying the phenotypic changes caused by the overexpression of ligand independent Notch 1 by using a tetracycline inducible promoter in an in vitro embryonic stem (ES) cells/OP9 stromal cells coculture system, recapitulating normal hematopoiesis. First, an in silico analysis of the promoters of Notch regulated genes (previously determined by microarray analysis) revealed that the motifs recognized by regulatory proteins known to mediate hematopoiesis were overrepresented. Notch 1 does not bind DNA but instead binds the CSL transcription factor to regulate gene expression. The in silico analysis also showed that there were putative CSL binding sites observed in the promoters of 28 out of 148 genes. A custom ChIP-chip array was used to assess the occupancy of CSL in the promoter regions of the Notch1 regulated genes in vivo and showed that 61 genes were bound by activated Notch responsive CSL. Then, comprehensive mapping of the CSL binding sites genome-wide using ChIP-seq analysis revealed that over 10,000 genes were bound within 10 kb of the TSS (transcription start site). The majority of the targets discovered by ChIP-seq belong to pathways that have been shown by others to crosstalk with Notch signaling. Finally, 83 miRNAs were significantly differentially expressed by greater than 1.5-fold during the course of in vitro hematopoiesis. Thirty one miRNA were up-regulated and fifty two were down-regulated. Overexpression of Notch1 altered this pattern of expression of microRNA: six miRNAs were up-regulated and four were down regulated as a result of activated Notch1 overexpression during the course of hematopoiesis. Time course analysis of hematopoietic development revealed that cells with Notch 1 overexpression mimic miRNA expression of cells in a less mature stage, which is consistent with our previous biological characterization.
Article
Aims: Although bone marrow cells are reported to migrate to the liver under circumstances of severe liver injury, the bone marrow cell type and the mechanisms in this process, remain to be clarified. We examined the involvement of hepatocyte growth factor (HGF) in this process and the cell type of migrated hematopoietic cells by HGF. Methods: The CD34(+) cells and colony forming cells in the peripheral blood were examined in HGF transgenic, recombinant HGF-administered, and HGF-expressing adenovirus-administered mice. The cell type mobilized by HGF was examined by the percentages of donor cells in the peripheral blood of the recipient mice transplanted with Lin(-)c-kit(+)Sca-1(+)CD34(+) cells and those with Lin(-)c-kit(+)Sca-1(+)CD34(-) cells. Expression of stem cell factor (SCF) was examined after the addition of HGF in MS-5 stromal cells. The numbers of the cells which were mobilized from bone marrow and recruited into liver by HGF were assessed using green fluorescence fluorescent (GFP)-chimera mice. Results: Mobilized CD34+ cells and colony forming cells in the peripheral blood were increased by HGF treatment. The cells mobilized by HGF were mostly Lin(-)c-kit(+)Sca-1(+)CD34(+) cells. Recruitment of bone marrow cells into liver was not suppressed in MMP-9-/- mice. Expression of SCF was induced by HGF in MS-5 stromal cells. However, expression of CXCR4, SDF-1, MMP-9 or VCAM-1 was not changed. The numbers of GFP-positive cells in liver 1 month after treatment by HGF was greater than that by G-CSF. Conclusion: The results of the present study suggest that HGF mobilizes and recruits hematopoietic progenitor cells from bone marrow into the liver through SCF-mediated mechanism.
Article
Full-text available
Here, we demonstrate a significant ex vivo expansion of human hematopoietic stem cells capable of repopulating in NOD/SCID mice. Using a combination of stem cell factor (SCF), Flk2/Flt3 ligand (FL), thrombopoietin (TPO), and a complex of IL-6 and soluble IL-6 receptor (IL-6/sIL-6R), we cultured cord blood CD34+ cells for 7 days and transplanted these cells into NOD/SCID mice. Bone marrow engraftment was judged successful when recipient animals contained measurable numbers of human CD45+ cells 10–12 weeks after transplantation. When cells were cultured with SCF+FL+TPO+IL-6/sIL-6R, 13 of 16 recipients were successfully engrafted, and CD45+ cells represented 11.5% of bone marrow cells in engrafted recipients. Cells cultured with a subset of these factors were less efficiently engrafted, both as measured by frequency of successful transplantations and prevalence of CD45+ cells. In animals receiving cells cultured with all 4 factors, human CD45+ cells represented various lineages, including a large number of CD34+ cells. The proportion of CD45+ cells in recipient marrow was 10 times higher in animals receiving these cultured cells than in those receiving comparable numbers of fresh CD34+ cells, and the expansion rate was estimated at 4.2-fold by a limiting dilution method. Addition of IL-3 to the cytokine combination abrogated the repopulating ability of the expanded cells. The present study may provide a novel culture method for the expansion of human transplantable hematopoietic stem cells suitable for clinical applications.
Article
Full-text available
To investigate the functional change of stromal cells along with differentiation, we used a differentiation-inducible mouse embryo fibroblast cell line, C3H10T1/2 (10T1/2). Stably determined preadipocyte and myoblast cell lines were established after a brief exposure of 10T1/2 cells to 5-azacytidine. These cell lines terminally differentiated into adipocytes and myotubes, respectively, under appropriate conditions. The hematopoiesis-supporting ability of each 10T1/2-derived cell line was examined by coculture with FACS-sorted murine hematopoietic stem cells (Thy-1lo c-kit+ Lin-). The number of granulocyte-macrophage progenitors (CFU-GM) was slightly reduced after 7 days of culture with parent 10T1/2 fibroblasts, whereas a marked increase in CFU-GM number was observed when the cells were cultured on preadipocytes. Mature adipocytes and myogenically determined cell lines, on the other hand, did not support CFU-GM growth. Further, Northern analysis showed that the preadipocyte cell line acquired the ability to produce a significant amount of stem cell factor (SCF), interleukin-6 (IL-6), leukemia inhibitory factor, and macrophage colony-stimulating factor mRNAs in response to IL-1 or lipopolysaccharide stimulation. Terminal adipocytic differentiation resulted in reduced ability to express these cytokine mRNAs. Similarly, highest IL-6 activity was detected in the supernatant of preadipocyte culture, whereas adipocytes did not secrete IL-6 even after IL-1 stimulation. Interestingly, hematopoiesis-nonsupporting myoblasts and myotubes also expressed abundant SCF mRNA, suggesting that SCF, per se, may not be sufficient for stem cell growth and survival. The 10T1/2-derived cell lines could provide a valuable tool to aid in the analysis of stromal cell development and the search for novel stromal factors.
Article
Full-text available
This report describes stroma-based and stroma-free cultures that maintain long-term engrafting hematopoietic cells for at least 14 days ex vivo. Umbilical cord blood (UCB) CD34(+) cells were cultured in transwells above AFT024 feeders with fetal-liver-tyrosine-kinase (FL) + stem cell factor (SCF) + interleukin 7 (IL-7), or FL + thrombopoietin (Tpo). CD34(+) progeny were transplanted into nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mice or preimmune fetal sheep. SCID repopulating cells (SRC) with multilineage differentiation potential were maintained in FL-SCF-IL-7 or FL-Tpo containing cultures for up to 28 days. Marrow from mice highly engrafted with uncultured or expanded cells induced multilineage human hematopoiesis in 50% of secondary but not tertiary recipients. Day 7 expanded cells engrafted primary, secondary, and tertiary fetal sheep. Day 14 expanded cells, although engrafting primary and to a lesser degree secondary fetal sheep, failed to engraft tertiary recipients. SRC that can be transferred to secondary recipients were maintained for at least 14 days in medium containing glycosaminoglycans and cytokines found in stromal supernatants. This is the first demonstration that ex vivo culture in stroma-noncontact and stroma-free cultures maintains "long-term" engrafting cells, defined by their capacity to engraft secondary or tertiary hosts. (Blood. 2001;97:3441-3449)
Article
To investigate the functional change of stromal cells along with differentiation, we used a differentiation-inducible mouse embryo fibroblast cell line, C3H10T1/2 (10T1/2). Stably determined preadipocyte and myoblast cell lines were established after a brief exposure of 10T1/2 cells to 5-azacytidine. These cell lines terminally differentiated into adipocytes and myotubes, respectively, under appropriate conditions. The hematopoiesis-supporting ability of each 10T1/2-derived cell line was examined by coculture with FACS-sorted murine hematopoietic stem cells (Thy-1lo c-kit+ Lin-). The number of granulocyte-macrophage progenitors (CFU-GM) was slightly reduced after 7 days of culture with parent 10T1/2 fibroblasts, whereas a marked increase in CFU-GM number was observed when the cells were cultured on preadipocytes. Mature adipocytes and myogenically determined cell lines, on the other hand, did not support CFU-GM growth. Further, Northern analysis showed that the preadipocyte cell line acquired the ability to produce a significant amount of stem cell factor (SCF), interleukin-6 (IL-6), leukemia inhibitory factor, and macrophage colony- stimulating factor mRNAs in response to IL-1 or lipopolysaccharide stimulation. Terminal adipocytic differentiation resulted in reduced ability to express these cytokine mRNAs. Similarly, highest IL-6 activity was detected in the supernatant of preadipocyte culture, whereas adipocytes did not secrete IL-6 even after IL-1 stimulation. Interestingly, hematopoiesis-nonsupporting myoblasts and myotubes also expressed abundant SCF mRNA, suggesting that SCF, per se, may not be sufficient for stem cell growth and survival. The 10T1/2-derived cell lines could provide a valuable tool to aid in the analysis of stromal cell development and the search for novel stromal factors.
Article
We established a differentiation-inducible preadipocyte cell line, designated A54 preadipocytes, from C3H10T1/2 (10T1/2) mouse embryo fibroblasts. A54 preadipocytes had marked hematopoiesis-supporting ability in vitro but this ability was lost after terminal differentiation to adipocytes. In this study, to identify molecules that contribute to the hematopoiesis-supporting ability of A54 preadipocytes, we screened genes that were differentially expressed in A54 preadipocytes and isolated seven novel genes by reverse transcriptase polymerase chain reaction mRNA differential display. An RNase protection assay confirmed that one of these genes was expressed at high levels in parent 10T1/2 cells and A54 preadipocytes but to a much lesser extent in fully differentiated A54 adipocytes. This gene was defined as a gene that was downregulated during adipocyte differentiation-1 (drad-1). The size of drad-1 mRNA was 8.2 kb, and the gene was expressed in other mouse preadipocytes, namely, ST2 and PA6 cells, that have hematopoiesis-supporting ability. Moreover, drad-1 was also found to be expressed in bone marrow in vivo. The function of the protein encoded by drad-1 is unknown, but the expression of the gene may be useful as a molecular marker of adipocyte differentiation.
Article
A liquid culture system is described whereby proliferation of haemopoietic stem cells (CFU-S), production of granulocyte precursor cells (CFU-C), and extensive granulopoiesis can be maintained in vitro for several months. Such cultures consist of adherent and non-adherent populations of cells. The adherent population contains phagocytic mononuclear cells, “epithelial” cells, and “giant fat” cells. The latter appear to be particularly important for stem cell maintenance and furthermore there is a strong tendency for maturing granulocytes to selectively cluster in and around areas of “giant fat” cell aggregations. By “feeding” the cultures at weekly intervals, between 10 to 15 “population doublings” of functionally normal CFU-S regularly occurs. Increased “population doublings” may be obtained by feeding twice weekly. The cultures show initially extensive granulopoiesis followed, in a majority of cases, by an accumulation of blast cells. Eventually both blast cells and granulocytes decline and the cultures contain predominantly phagocytic mononuclear cells. Culturing at 33°C leads to the development of a more profuse growth of adherent cells and these cultures show better maintenance of stem cells and increased cell density. When tested for colony stimulating activity (CSA) the cultures were uniformly negative. Addition of exogenous CSA caused a rapid decline in stem cells, reduced granulopoiesis and an accumulation of phagocytic mononuclear cells.
Article
An efficient system was developed that induced the differentiation of embryonic stem (ES) cells into blood cells of erythroid, myeloid, and B cell lineages by coculture with the stromal cell line OP9. This cell line does not express functional macrophage colony-stimulating factor (M-CSF). The presence of M-CSF had inhibitory effects on the differentiation of ES cells to blood cells other than macrophages. Embryoid body formation or addition of exogenous growth factors was not required, and differentiation was highly reproducible even after the selection of ES cells with the antibiotic G418. Combined with the ability to genetically manipulate ES cells, this system will facilitate the study of molecular mechanisms involved in development and differentiation of hematopoietic cells.
Article
The regulation of hematopoiesis has been suggested to take place in close association with various cell types found in the bone marrow (BM) microenvironment. In the present study the role of fibroblasts, adipocytes and cell surface heparan sulfate in regulating hematopoiesis in an in vitro mouse system was examined. Mouse BM cells were allowed to adhere to a mouse embryo fibroblast cell line (C3H 10T1/2) or a clonally derived adipogenically determined derivative (Clone D) of the 10T1/2 cell line. Nonadherent cells were removed, cultures were overlaid with semisolid media supplemented with growth factors and colony formation by granulocyte-macrophage (CFU-GM), erythroid (BFU-E) and multipotential (CFU-GEMM) progenitor cells was quantitated. Adherence and co-culture of BM cells with the fibroblast cell line resulted in increased numbers of total CFU-GM and CFU-GEMM colonies. In contrast, adherence and co-culture of BM cells with the adipocytic cell line resulted in an increase only in CFU-GEMM colonies. Morphological analysis revealed a preferential adherence/growth of granulocyte and macrophage progenitors at the expense of bipotent granulocyte-macrophage progenitors to the fibroblastic cell line and an increase in the adherence/growth of granulocyte progenitors to the adipogenic cell line. Progenitor cell adherence was abolished when the fibroblastic or adipocytic cell lines were pretreated with heparitinase. These results demonstrate enhanced proliferation/differentiation of hematopoietic progenitor cells when there is direct contact between hematopoietic progenitors and cell types characteristic of those found in the microenvironment and that heparan sulfate and different types of stromal cells appear to play different roles in this interaction.
Article
During mouse embryogenesis the production of "primitive" erythrocytes (EryP) precedes the production of "definitive" erythrocytes (EryD) in parallel with the transition of the hematopoietic site from the yolk sac to the fetal liver. On a macrophage colony-stimulating factor-deficient stromal cell line OP9, mouse embryonic stem cells were shown to give rise to EryP and EryD sequentially with a time course similar to that seen in murine ontogeny. Studies of the different growth factor requirements and limiting dilution analysis of precursor frequencies indicate that most EryP and EryD probably developed from different precursors by way of distinct differentiation pathways.