ArticlePDF Available

A Rapid and Efficient Method for Expansion of Human Mesenchymal Stem Cells

Authors:

Abstract and Figures

During the past decade, there has been much interest in the use of human mesenchymal stem cells (hMSCs) in bone tissue engineering. HMSCs can be obtained relatively easily and expanded rapidly in culture, but for clinical purposes large numbers are often needed and the cost should be kept to a minimum. A rapid and efficient culturing protocol would therefore be beneficial. In this study, we examined the effect of different medium compositions on the expansion and osteogenic differentiation of bone marrow-derived hMSCs from 19 donors. We also investigated the effect of low seeding density and dexamethasone on both hMSCs expansion and their in vitro and in vivo osteogenic differentiation capacity. HMSCs seeded at a density of 100 cells/cm2 had a significantly higher growth rate than at 5000 cell/cm2, which was further improved by the addition of dexamethasone. Expanded hMSCs were characterized in vitro on the basis of positive staining for CD29, CD44, CD105, and CD166. The in vitro osteogenic potential of expanded hMSCs was assessed by flow cytometric staining for alkaline phosphatase. In vivo bone-forming potential of the hMSCs was assessed by seeding the cells in ceramic scaffolds, followed by subcutaneous implantation in nude mice and histopathologic assessment of de novo bone formation after 6-week implantation. Expanded hMSCs from all donors displayed similar osteogenic potential independent of the culture conditions. On the basis of these results we have developed an efficient method to culture hMSCs by seeding the cells at 100 cells/cm2 in an alpha-minimal essential medium-based medium containing dexamethasone.
Content may be subject to copyright.
A Rapid and Efficient Method for Expansion of Human
Mesenchymal Stem Cells
SANNE K. BOTH, B.Sc.,
1
ADRIE J.C. VAN DER MUIJSENBERG, B.Sc.,
1
CLEMENS A. VAN BLITTERSWIJK, Ph.D.,
1
JAN DE BOER, Ph.D.,
1
and JOOST D. DE BRUIJN, Ph.D.
2
ABSTRACT
During the past decade, there has been much interest in the use of human mesenchymal stem cells (hMSCs)
in bone tissue engineering. HMSCs can be obtained relatively easily and expanded rapidly in culture, but
for clinical purposes large numbers are often needed and the cost should be kept to a minimum. A rapid
and efficient culturing protocol would therefore be beneficial. In this study, we examined the effect of
different medium compositions on the expansion and osteogenic differentiation of bone marrow–derived
hMSCs from 19 donors. We also investigated the effect of low seeding density and dexamethasone on both
hMSCs expansion and their in vitro and in vivo osteogenic differentiation capacity. HMSCs seeded at a
density of 100 cells/cm
2
had a significantly higher growth rate than at 5000 cell/cm
2
, which was further
improved by the addition of dexamethasone. Expanded hMSCs were characterized in vitro on the basis
of positive staining for CD29, CD44, CD105, and CD166. The in vitro osteogenic potential of expanded
hMSCs was assessed by flow cytometric staining for alkaline phosphatase. In vivo bone-forming potential
of the hMSCs was assessed by seeding the cells in ceramic scaffolds, followed by subcutaneous implan-
tation in nude mice and histopathologic assessment of de novo bone formation after 6-week implantation.
Expanded hMSCs from all donors displayed similar osteogenic potential independent of the culture con-
ditions. On the basis of these results we have developed an efficient method to culture hMSCs by seeding
the cells at 100 cells/cm
2
in an a-minimal essential medium–based medium containing dexamethasone.
INTRODUCTION
THE CURRENT AND MOST FREQUENTLY USED APPROACH for
the treatment of large bone defects is autologous or
allogeneic bone grafting. Both methods have distinct draw-
backs, including donor site pain and limited availability for
autograft, and the risk of immune reaction and disease trans-
mission for allograft. The creation of a tissue-engineered
implant, which consists of a suitable biomaterial scaffold
covered with the patient’s own osteogenic cells, could pro-
vide an alternative approach.
Human bone marrow contains a multipotent cell popu-
lation, which has the ability to differentiate into several
mesenchymal lineages,
1
including bone, cartilage, fat, and
muscle.
2
These cells were initially isolated by their ad-
herence to tissue culture surfaces
3,4
and are referred to as
colony-forming unit fibroblasts, marrow stromal cells, or
mesenchymal stem cells. Because of their multipotent na-
ture, these cells have attracted much interest for use in
cell and gene therapy, as well as for treating large bone
defects.
5,6
When grown under osteogenic differentiation
conditions, it has been shown that human mesenchymal
stem cells (hMSCs) not only possess in vitro osteogenic
potential
7,8
but also can form bone tissue in vivo, as dem-
onstrated by subcutaneous implantation in immunodeficient
mice.
9–11
1
Institute for Biomedical Technology, University of Twente, Bilthoven, the Netherlands.
2
Department of Materials, Queen Mary University of London, London, United Kingdom.
TISSUE ENGINEERING
Volume 13, Number 1, 2007
#Mary Ann Liebert, Inc.
DOI: 10.1089/ten.2005.0513
3
Several markers have been suggested for the identifica-
tion and osteogenic differentiation of hMSCs.
10–14
The
hMSCs are positive for the surface expressed antigen en-
doglin receptors CD105 and CD44, the activated leukocyte
cell adhesion molecule CD166, and b-integrin CD29.
12
Developmental markers such as alkaline phosphatase (ALP)
and STRO-1 can be used to characterize osteogenic and un-
differentiated hMSCs, respectively.
13–15
Several culture medium supplements that affect the ex-
pansion and differentiation of hMSCs have been reported.
For instance, basic fibroblastic growth factor and lithium
can enhance the proliferation of hMSCs.
15,16
The addition
of the synthetic glucocorticoid dexamethasone to the me-
dium will induce hMSCs into the osteogenic lineage, thereby
changing the shape of the cells
17
and increasing the ex-
pression of osteoblast markers such as ALP.
7,18,19
Although in vitro osteogenic differentiation of hMSCs and
proof of principle for the creation of an autologous tissue-
engineered implant has been shown,
9,20,21
we still lack a
well-defined protocol for the isolation and expansion of these
cells to make this technique available for clinical purposes.
Surgical skills, volume of the aspirated bone marrow cells,
22
and the donor age and variation in growth rate between dif-
ferent donors significantly influence the creation of a suc-
cessful living implant.
9,20,21
Besides commitment of hMSCs
to the osteogenic lineage, large numbers of cells are needed
for creating a tissue-engineered implant that can fill large
bone defects. Since expansion of hMSCs is labor intensive
and therefore expensive, the use of an optimal culture system
is desirable. For example, on the basis of using a scaffold
containing 8 million cells/mL,
23
20 mL of tissue-engineered
implant used for spinal fusion surgery would require about
160 million cells. By using previously established proto-
cols,
9,19,24
it would take 15–29 days and approximately 24
person-hours to reach enough cells. Although this protocol
was previously used to create tissue-engineered living bone
equivalents, which were also used in a clinical trial (data
submitted separately to PLoS Medicine, December 2006), it
was time consuming and labor intensive because of the
amount of subculturing needed. There is therefore a need
for an optimal protocol for expansion and osteogenic differ-
entiation of hMSCs.
20
In this report, we describe a method for
the expansion of osteogenic hMSCs under optimized con-
ditions to allow treatment of bone defects.
MATERIALS AND METHODS
Harvest and culture of hMSCs
Bone marrow aspirates were obtained from 19 donors who
were undergoing total hip replacement surgery and had given
informed consent (donor information is provided in Table 1).
Nucleated cells were counted in the aspirate and plated at a
density of 500,000 cells/cm
2
in a-minimal essential medium
(aMEM) proliferation medium (see below; donors 1–15) or
aMEM osteogenic medium (see below; donors 11–15). The
hMSCs from donors 16–19 were harvested and proliferated
as described above and then cryopreserved. After defrosting,
TABLE 1. INFORMATION ON DONORS ,EXPERIMENTS, AND NUMBER OF DAYS NEEDED FOR HUMAN MESENCHYMAL STEM CELLS
TO REACH 20010
6
CELLS WHEN SEEDED AT 100 AND 5000 CELLS/CM
2
MEM
versus
100 cells/cm
2
versus 5000 þ/
Days needed to reach
20010
6
cells
Donor Aspiration site Sex Age DMEM cells/cm
2
Dexamethasone In vivo 100 cells 5000 cells
1 Crista F 72 HH15 18
2 Crista F 87 HH19 24
3 Crista F 61 HH21 29
4 Crista F 63 HH16 17
5 Crista F 69 HH19 23
6 Acetabulum M 29 HH12 15
7 Acetabulum F 63 HH15 19
8 Acetabulum M 56 HH12 19
9 Iliac crest F 28 HH14 20
10 Acetabulum F 25 HH13 16
11 Crista F 77 HHH14 15
12 Acetabulum F 44 HHH16 18
13 Iliac crest F 26 HHH14 16
14 Acetabulum M 48 HHH16 18
15 Iliac crest M 56 HHH16 19
16 Acetabulum F 28 H
17 Acetabulum M 49 H
18 Iliac Crest M 42 H
19 Iliac crest F 36 H
Abbreviations: aMEM, a-minimal essential medium; DMEM, Delbecco’s modified Eagle medium; F, female; M, male.
4BOTH ET AL.
they were plated at 5000 cells/cm
2
and grown in the different
culture media as described below (n¼5 for each donor).
The aMEM proliferation medium contained minimal es-
sential medium (GIBCO, Carlsbad, CA), 10% fetal bovine
serum of a selected batch (FBS; BioWhittaker, Australia,
lot: 0S025F); 0.2 mM L-ascorbic acid-2-phosphate (Sigma,
St. Louis, MO), penicillin G (100 U/mL, Invitrogen, Carls-
bad, CA); streptomycin (100 mg/mL, Invitrogen); 2 mM L-
glutamine (Sigma) and 1ng/mL basic fibroblast growth
factor (Instruchemie, Delfzijl, the Netherlands). The aMEM
osteogenic medium was composed of the proliferation me-
dium supplemented with 10 nM dexamethasone (Sigma).
Delbecco’s modified Eagle medium (DMEM) proliferation
medium comprised 10% FBS (BioWhittaker), 1% nones-
sential amino acids 100 (Sigma), penicillin G (100 U/mL;
Invitrogen), streptomycin (100 mg/mL; Invitrogen), 2 mM L-
glutamine (Sigma), 1 mM sodium pyruvate (GIBCO), 1 mM
Hepes (GIBCO) and 1 ng/mL basic fibroblast growth factor
(Instruchemie). DMEM osteogenic medium was composed
of DMEM proliferation medium supplemented with 10 nM
dexamethasone (Sigma). The batch of FBS selected for the
hMSCs in this study was tested in vitro for cell growth,
expression of ALP, by FACS and the enzymatic expression
of ALP and in vivo for bone formation of the hMSCs. The
cells were grown at 37
8
C in a humidified atmosphere of 5%
carbon dioxide. Cultures were refreshed twice a week. When
the cells reached near confluence (80–90%), they were wa-
shed with phosphate-buffered saline (PBS; Sigma), detached
with 0.05% trypsin (GIBCO), containing 1 mM ethylene-
diaminetetraacetic acid and subcultured.
Flow cytometry
For flow cytometry, hMSCs from donors 1–15 were
detached and centrifuged at 100 gfor 10 min at 48C. The
cell pellets were resuspended in PBS containing 5% bovine
serum albumin (Sigma) and 0.05% sodium azide (NaN
3
;
Sigma) and stored on ice for 15 min. Next, the cells were
incubated in block buffer containing control mouse anti-
human IgG2a (Dako, Glostrup, Denmark); anti-ALP anti-
body (hybridoma B4-78), anti-CD29 monoclonal antibody
(Dako), anti-CD44 (Pharming, Leiden, the Netherlands)
antibody, anti-CD105 (Dako) antibody, or anti-CD166
(RDI, Concord, MA) monoclonal antibody. The cells were
incubated on ice for 45 min and washed three times with
PBS containing 1% BSA and 0.05% NaN
3
(wash buffer).
Incubation with the secondary antibody, goat anti-mouse-
IgG–fluorescein isothiocyanate (Dako), was performed
for 45 min on ice. After the hMSCs were washed 3 times
with wash buffer, the cells were incubated for 10 min with
10 mL Viaprobe (Becton Dickinson, Franklin Lakes, NJ) for
live-dead staining. The cell suspension was analyzed by us-
ing a flow cytometer (Becton Dickinson). From each sam-
ple, 10,000 events were collected in duplicate and the data
were analyzed via Cell Quest software (Becton Dickinson).
For statistical analysis of the percentages of positive cells
of the donors, a Student t-test was performed; significance
was determined at a pvalue less than .05.
ALP activity
The enzyme activity of ALP was measured in triplicate
for donors 16–19 by washing the cells with PBS and in-
cubating them for 10–15 minutes in a 20 mM paranitro-
phenol solution (Sigma) in 1 M diethanolamine (Sigma),
1 mM magnesium chloride (Sigma). The activity of the ALP
enzyme was measured with a microplate reader (BIOTEC
Instruments, Switzerland) at 310 nm and normalized for cell
number.
In vivo implantation
To assess the in vivo bone-forming capacity of various
passages of hMSCs from donors 1–15, they were seeded
onto porous biphasic calcium phosphate scaffolds (2–4 mm)
and implanted in nude mice. The hMSCs were seeded by
adding a 100-mL drop cell suspension, containing 200,000
cells, on each biphasic calcium phosphate scaffold (n¼6 per
experiment). After 2 h of adherence, the medium was added
and scaffolds were cultured for 7 days in aMEM osteogenic
medium. Before implantation, the particles were placed in
serum-free medium for approximately 1–2 h and washed 3
times with PBS. Immunodeficient mice (HsdCpb:NMRI-nu,
Harlan) were anesthetized by an intramuscular injection of
0.05 mL ketamine, xylazine, and atropine mixture. Before
surgery, the skin on both lateral sites of the spine was
cleaned with 70% alcohol, and 6 subcutaneous pockets were
created in each mouse. In each pocket, 3 tissue-engineered
samples were inserted and each culture condition was di-
vided over three mice. Six weeks postoperatively, the ani-
mals were euthanized by carbon dioxide. The explanted
samples were fixated in 1.5% glutaraldehyde (Sigma) in 0.14
M cacodylic acid buffer, dehydrated by increasing ethanol
steps, and embedded in methylmethacrylate (Sigma). Sec-
tions were made by using a diamond saw (Leica SP1600,
Wetzlar, Germany) and stained with 1% methylene blue
(Sigma) and 0.3% basic fuchsin solution (Sigma).
25
RESULTS
Effect of culture medium on hMSC growth
The cells of donors 16–19 were cultured for 2 passages
in the 2 different basal media. When hMSCs were proli-
ferated in aMEM for 4 days, the number of cells obtained
was significantly higher for all 4 donors compared with
hMSC proliferated in a DMEM proliferation medium,
regardless of the passage number (n¼4 for each donor,
P<.05) (Fig. 1A).
To investigate the effect of different media on the oste-
ogenic potential of hMSCs, the cells were cultured for 7
days in osteogenic aMEM-based and DMEM-based media.
EXPANSION OF MESENCHYMAL STEM CELLS 5
When grown in aMEM-based medium, all 4 donors (16–
19) displayed increased ALP enzyme activity when dexa-
methasone had been added (differentiation medium). In
DMEM-based medium, this difference was less apparent
(Fig. 1B). In addition, overall ALP enzyme activity was
higher when the cells were cultured in aMEM medium as
compared with DMEM.
Effect of seeding density and dexamethasone
on hMSC proliferation
With all 15 donors tested, the cultures seeded at a density
of 100 cells/cm
2
reached 200 million cells within 12–21
days of passage one (Fig. 1). In contrast, hMSCs seeded at
5000 cells/cm
2
, as described in our previous culturing
protocol,
9,19
had to be subcultured for at least 2 passages
(three passages for most donors) and cultured for 15–29
days before 200 million cells were reached (Fig. 2, Table 1).
By using the lower seeding density, the cultures there-
fore reached 200 million cells approximately 4.1 days
earlier compared with cells seeded at 5000 cells/cm
2
. Dexa-
methasone stimulates differentiation of hMSCs, and some
hMSCs culture protocols use it throughout the entire culture
period.
26
Therefore, we investigated the effect of dexa-
methasone on hMSC proliferation by culturing them in
proliferation medium or osteogenic medium throughout the
entire proliferation period. The number of cells obtained
from the primary culture was higher for all five donors when
grown in osteogenic medium compared with proliferation
medium and also at later passages independent of the
seeding density (Fig. 3). Furthermore, dexamethasone ap-
peared to stimulate hMSC proliferation at later passages as
well.
Osteogenic differentiation
Cells grown at 100 cells/cm
2
in the presence of dexa-
methasone resulted in the fastest and most labor-efficient
method to expand hMSCs. To confirm that hMSCs expanded
by using this protocol retained their osteogenic potential, we
first analyzed the effect of seeding density and dexameth-
asone on the hMSC phenotype with a panel of hMSC
markers. The hMSC markers CD29, CD44, CD105, and
CD166 were expressed in hMSCs of all donors (1–15) in all
culture conditions, and the expression varied between 95%
and 100% (Fig. 4). To investigate whether the culture con-
ditions influenced the osteogenic potential of the hMSCs,
we measured expression of the osteogenic marker ALP.
From previous experiments (data not shown), we deter-
mined that after approximately 7 days of culture in the
presence of dexamethasone, ALP expression of the cells
reached its optimum. To assess whether the continuous
presence of dexamethasone influenced the osteogenic po-
tential of the hMSCs, cells previously cultured without
dexamethasone were also cultured with dexamethasone for
Number of cells (log)
1E+13
1E+12
1E+11
1E+10
1E+09
1E+08
10000000
10000000 5 10 15
Days
20 25 30 35
5000 cells
5000 cells + dex
100 cells
100 cells + dex
FIG. 2. The effect of seeding density and culture medium on
human mesenchymal stem cell proliferation. Progressive cell
numbers are expressed against culture time. The cells were seeded
at 5000 or 100 cells/cm
2
in proliferation and osteogenic media
(n¼5 for each donor). Error bars for each passage were too small
to be visualized in the graph. Because of variants in proliferation
rate between donors, a representative graph of the growth curve of
donor 2 is given. dex: dexamethasone.
1e+6
8e+6
6e+6
Amount of cells
4e+6
2e+6
0Donor 16 Donor 17 Donor 18 Donor 19
DMEM
Alpha MEM A B
DMEM
Alpha MEM
Donor 16 Donor 17 Donor 18 Donor 19
6
5
4
3
2
1
0
Fold induction of ALP
FIG. 1. (A) Proliferation of human mesenchymal stem cells (hMSCs) in a-minimal essential medium (aMEM) versus Delbecco’s
modified Eagle medium (DMEM). When hMSCs were proliferated in aMEM for 4 days, the number of cells obtained was significantly
higher for all 4 donors compared with hMSCs proliferated in a DMEM proliferation medium, regardless of the passage number (n¼4;
P<.05). (B) Effect of aMEM or DMEM medium on alkaline phosphate (ALP) enzyme activity. The hMSCs were proliferated in
aMEM and DMEM proliferation and differentiation media, and the ALP enzyme activity was measured as activity per cell. The fold
increase in ALP enzyme activity of the proliferation versus osteogenic medium is displayed in the graph.
6BOTH ET AL.
7 days. At both seeding densities, ALP expression signifi-
cantly increased when the cells were cultured in the pres-
ence of dexamethasone compared with cells cultured in
proliferation medium (Figure 3). ALP induction was sim-
ilar between cells seeded at different densities and those
with comparable population doublings. The induction of
ALP by dexamethasone did not significantly, regardless of
whether hMSCs had been cultured in previous passages in
the presence of dexamethasone or only for 7 days.
In vivo bone-forming potential of hMSCs
After 6 weeks of implantation, the samples of all donors
exhibited direct deposition of bone tissue, irrespective of
seeding density or presence of dexamethasone in the cul-
ture medium. The newly formed bone comprised osteocytes
embedded within the bone matrix and layers of osteoblasts
(Fig. 5). Besides fibrous tissue and newly formed bone, no
other connective tissue formation was observed with any of
the donors. Microscopic observation was performed blinded
by 3 independent observers, who each examined 5–8 slides
of each implanted condition. Between the different donors
the amount of newly formed bone varied, but not within the
different culture conditions from a single donor.
DISCUSSION
In this study, we demonstrate that the use of an aMEM-
based medium is superior for both expansion and the
osteogenic differentiation of hMSCs, as compared with a
DMEM-based medium. The expansion of hMSCs at low
density results in a higher proliferation rate compared with
cells proliferated at a higher density, which is further stim-
ulated by the addition of dexamethasone. Neither seeding
densities nor the addition of dexamethasone interferes with
the osteogenic in vitro and in vivo potential of the hMSCs.
The use of aMEM or DMEM medium for the expansion
of hMSCs has been investigated before by Jaiswal et al.
7
Interestingly, they found that fold increase of ALP enzyme
activity was much higher with DMEM culture, a finding in
contrast with our results. Moreover, they also stated that
mineralization was sparsely detected in hMSCs cultured in
aMEM osteogenic medium, which also conflicts with pre-
vious results.
27
In this paper, we report that hMSCs pro-
liferate faster at lower seeding densities. To investigate the
optimal density, we tested the growth rate of the hMSCs
also at densities lower than 100 cells/cm
2
and concluded that
the growth rate at those lower densities was even higher
then when seeded at 100 cells/cm
2
(data not shown). Un-
fortunately, at these low densities the cells form a few iso-
lated cell colonies, which do not fill up the entire culture
flask. Therefore, we concluded that seeding 100 cells/cm
2
represents the most efficient expansion. To create an even
better expansion environment for the hMSCs, it might be
beneficial to refresh not the entire culture media but part of
it. With refreshing the entire culture, media components
secreted by the cells will be removed and the cells will have
to adept to the new environment. When cells are seeded at
low density, they display a lag phase of 3–5 days before
going into a rapid exceptional growth phase. At the end
of the lag phase it has been reported that the cells secrete
5000 cells 100 cells
100
80
60
%positive cells
40
20
0
control medium
7 days + dex
entire culture + dex
FIG. 3. The effect of seeding density and culture medium on the
expression of alkaline phosphate (ALP) of human mesenchymal
stem cells (hMSCs). The percentage of positive cells expressing
ALP of donors 6–11 were measured after proliferating the cells at
5000 and 100 cells/cm
2
. Cells proliferated without dexamethasone
(dex) were cultured without (A) and with (B) dexamethasone for
7 days, or they were continuously cultured in the presence of
dexamethasone (C). ALP activity increased significantly for all 6
donors when dexamethasone was present in the media, regardless
of the proliferation medium previously used (P<.05). Prolifer-
ating hMSCs with or without dexamethasone had no significant
influence on the ALP expression; the seeding densities also had no
significant effect.
120
100
80
60
%positive cells
40
20
0
CD29 CD44 CD109 CD166
100 cells/cm2
5000 cells/cm2
FIG. 4. The effect of seeding density on the expression of the
human mesenchymal stem cell (hMSC)–related markers CD29,
CD44, CD105, and CD166. Cells of donor 7 were cultured at a
seeding density of 100 cells/cm
2
and 5000 cells/cm
2
in prolifer-
ation medium until 200 million cells was reached.
EXPANSION OF MESENCHYMAL STEM CELLS 7
high levels of dickkopf-1 (DKK-1), inhibitor of the Wnt-
signaling pathway.
28
High levels of DKK-1 allow the hMSCs
to re-enter the cell cycle and therefore be responsible for
the rapid growth. However, these data contradict the results
of de Boer et al.
16
which demonstrate that the addition
of lithium, a stimulant of the Wnt-signaling pathway, in-
creases the expansion rate of hMSCs. No real conclusion
can be made as to what causes the cells to grow at a higher
rate when seeded at low densities; it is very likely that
several different factors contribute to this phenomenon.
The effect of increased expansion of hMSCs cultured at
lower densities has also been reported by Colter et al.
29
They reported that there might be a minor population of
small agranular cells, also referred to as recycling stem
cells, that give rise to a new population of small densely
granular cells and are thereby responsible for the rapid
expansion when hMSCs are seeded at lower densities. They
also reported that in replated low-density passages, their
hMSCs retained their ability to generate single cell–derived
colonies and thereby retained their multipotentiality for
differentiation. This is in contrast with an observation we
made. We observed that the osteogenic potential of the
cells both in vitro and in vivo seemed to be decreasing with
repeated passages of the hMSCs, indicating that the cells
might lose their multipotentiality for differentiation. This
change can be observed in vitro by flow cytometry. It has
been reported by Mendes et al.
18
that an increase in ALP
induction by dexamethasone is related to the occurrence of
bone formation in vivo. The addition of dexamethasone to
the medium is not a new concept. Rat MSC cultures are
always expanded in the presence of dexamethasone,
26,30
but for hMSCs dexamethasone is almost only used for dif-
ferentiation of the cells, not for expansion. We clearly
show herein that dexamethasone does not only influ-
ence the osteogenic potential of hMSCs but lso enhances
their proliferation rate, without interfering with the in vivo
bone-forming capacities of the hMSCs.
In conclusion, these results indicate that the optimal way
to rapidly expand hMSCs is by seeding the cells at a density
of 100 cells/cm
2
in an aMEM-based medium containing
dexamethasone.
ACKNOWLEDGMENT
The authors would like to acknowledge the orthopedic
departments from the University Hospital of Utrecht, Ac-
ademic Hospital of Maastricht, and the Sint Maartens clinic
in Nijmegen for providing bone marrow aspirates for this
study.
REFERENCES
1. Owen, M., and Friedenstein, A.J. Stromal stem cells: marrow-
derived osteogenic precursors. Ciba Found Symp 136, 42,
1988.
2. Pittenger, M.F., Mackay, A.M., Beck, S.C., Jaiswal, R.K.,
Douglas, R., Mosca, J.D., Moorman, M.A., Simonetti, D.W.,
Craig, S., and Marshak, D.R. Multilineage potential of adult
human mesenchymal stem cells. Science 284, 143, 1999.
3. Friedenstein, A.J., Gorskaja, J.F., and Kulagina, N.N. Fibro-
blast precursors in normal and irradiated mouse hematopoi-
etic organs. Exp Hematol 4, 267, 1976.
4. Friedenstein, A.J., Chailakhyan, R.K., and Gerasimov, U.V.
Bone marrow osteogenic stem cells: in vitro cultivation and
transplantation in diffusion chambers. Cell Tissue Kinet 20,
263, 1987.
5. Prockop, D.J. Marrow stromal cells as stem cells for nonhe-
matopoietic tissues. Science 276, 71, 1997.
FIG. 5. In vivo bone formation by human mesenchymal stem
cells (hMSCs). Bone was formed on porous biphasic calcium phos-
phate particles after 6 weeks of subcutaneous implantation in nude
mice. The newly formed bone (b) was deposited against the ma-
terial (CaP) surfaces (A–C). In some samples, an abundant amount
of newly formed bone was present. Embedded osteocytes (small
arrow) and a layer of osteoblasts (big arrow) were observed within
the newly formed bone. Irrespective of culture medium or cul-
ture method, hMSCs from all donors (1–15) exhibited in vivo
bone formation. Color images available online at www.liebertpub
.com /ten.
8BOTH ET AL.
6. Horwitz, E.M., Prockop, D.J., Fitzpatrick, L.A., Koo, W.W.,
Gordon, P.L., Neel, M., Sussman, M., Orchard, P., Marx, J.C.,
Pyeritz, R.E., and Brenner, M.K. Transplantability and ther-
apeutic effects of bone marrow-derived mesenchymal cells
in children with osteogenesis imperfecta. Nat Med 5, 309,
1999.
7. Jaiswal, N., Haynesworth, S.E., Caplan, A.I., and Bruder, S.P.
Osteogenic differentiation of purified, culture-expanded hu-
man mesenchymal stem cells in vitro. J Cell Biochem 64,
295, 1997.
8. Haynesworth, S.E., Goshima, J., Goldberg, V.M., and Caplan,
A.I. Characterization of cells with osteogenic potential from
human marrow. Bone 13, 81, 1992.
9. Mendes, S.C., Van Den Brink, I., De Bruijn, J.D., and Van
Blitterswijk, C.A. In vivo bone formation by human bone
marrow cells: effect of osteogenic culture supplements and
cell densities. J Mater Sci Mater Med 9, 855, 1998.
10. Arinzeh, T.L., Tran, T., McAlary, J., and Daculsi, G. A
comparative study of biphasic calcium phosphate ceramics
for human mesenchymal stem-cell-induced bone formation.
Biomaterials 26, 3631, 2005.
11. Kasten, P., Vogel, J., Luginbuhl, R., Niemeyer, P., Tonak, M.,
Lorenz, H., Helbig, L., Weiss, S., Fellenberg, J., Leo, A.,
Simank, H.G., and Richter, W. Ectopic bone formation as-
sociated with mesenchymal stem cells in a resorbable calcium
deficient hydroxyapatite carrier. Biomaterials 26, 5879, 2005.
12. Le Blanc, K., Tammik, L., Sundberg, B., Haynesworth, S.E.,
and Ringden, O. Mesenchymal stem cells inhibit and stimu-
late mixed lymphocyte cultures and mitogenic responses in-
dependently of the major histocompatibility complex. Scand J
Immunol 57, 11, 2003.
13. Oyajobi, B.O., Lomri, A., Hott, M., and Marie, P.J. Isolation
and characterization of human clonogenic osteoblast pro-
genitors immunoselected from fetal bone marrow stroma us-
ing STRO-1 monoclonal antibody. J Bone Miner Res 14, 351,
1999.
14. Stewart, K., Walsh, S., Screen, J., Jefferiss, C.M., Chainey, J.,
Jordan, G.R., and Beresford, J.N. Further characterization of
cells expressing STRO-1 in cultures of adult human bone
marrow stromal cells. J Bone Miner Res 14, 1345, 1999.
15. Walsh, S., Jefferiss, C., Stewart, K., Jordan, G.R., Screen, J.,
and Beresford, J.N. Expression of the developmental markers
STRO-1 and alkaline phosphatase in cultures of human mar-
row stromal cells: regulation by fibroblast growth factor
(FGF)-2 and relationship to the expression of FGF receptors
1-4. Bone 27, 185, 2000.
16. De Boer, J., Wang, H.J., and Van Blitterswijk, C. Effects of
Wnt signaling on proliferation and differentiation of human
mesenchymal stem cells. Tissue Eng 10, 393, 2004.
17. Byers, R.J., Brown, J., Brandwood, C., Wood, P., Staley, W.,
Hainey, L., Freemont, A.J., and Hoyland, J.A. Osteoblastic
differentiation and mRNA analysis of STRO-1-positive hu-
man bone marrow stromal cells using primary in vitro culture
and poly (A) PCR. J Pathol 187, 374, 1999.
18. Cheng, S.L., Yang, J.W., Rifas, L., Zhang, S.F., and Avioli,
L.V. Differentiation of human bone marrow osteogenic stro-
mal cells in vitro: induction of the osteoblast phenotype by
dexamethasone. Endocrinology 134, 277, 1994.
19. Mendes, S.C., Tibbe, J.M., Veenhof, M., Both, S., Oner, F.C.,
van Blitterswijk, C.A., and de Bruijn, J.D. Relation between
in vitro and in vivo osteogenic potential of cultured human
bone marrow stromal cells. J Mater Sci Mater Med 15, 1123,
2004.
20. Phinney, D.G., Kopen, G., Righter, W., Webster, S., Tremain,
N., and Prockop, D.J. Donor variation in the growth proper-
ties and osteogenic potential of human marrow stromal cells.
J Cell Biochem 75, 424, 1999.
21. D’Ippolito, G., Schiller, P.C., Ricordi, C., Roos, B.A., and
Howard, G.A. Age-related osteogenic potential of mesen-
chymal stromal stem cells from human vertebral bone mar-
row. J Bone Miner Res 14, 1115, 1999.
22. Muschler, G.F., Boehm, C., and Easley, K. Aspiration to
obtain osteoblast progenitor cells from human bone marrow;
the influence of aspiration volume. J Bone Miner Res 79,
1699, 1997.
23. Kruyt, M.C., de Bruijn, J.D., Yuan, H., van Blitterswijk, C.A.,
Verbout, A.J., Oner, F.C., and Dhert, W.J. Optimization of
bone tissue engineering in goats: a perioperative seeding
method using cryopreserved cells and localized bone forma-
tion in calcium phosphate scaffolds. Transplantation 77, 359,
2004.
24. Kuznetsov, S.A., Krebsbach, P.H., Satomura, K., Kerr, J.,
Riminucci, M., Benayahu, D., and Robey, P.G. Single-colony
derived strains of human marrow stromal fibroblasts form
bone after transplantation in vivo. J Bone Miner Res 12, 1335,
1997.
25. van der Lubbe, H.B., Klein, C.P., and de Groot, K. A simple
method for preparing thin (10 microM) histological sections
of undecalcified plastic embedded bone with implants. Stain
Technol 63, 171, 1988.
26. Maniatopoulos, C., Sodek, J., and Melcher, A.H. Bone for-
mation in vitro by stromal cells obtained from bone marrow
of young adult rats. Cell Tissue Res 254, 317, 1988.
27. De Boer, J., Siddappa, R., Gaspar, C., Van Aapeldoorn, A.,
Fodde, R., and Van Blitterswijk, C. Wnt signaling inhibits
osteogenic differentiation of human mesenchymal stem cells.
Bone 34, 818, 2004.
28. Gregory, C.A., Singh, H., Perry, A.S., and Prockop, D.J. The
Wnt signaling inhibitor dickkopf-1 is required for reentry into
the cell cycle of human adult stem cells from bone marrow.
J Biol Chem 278, 28067, 2003.
29. Colter, D.C., Class, R., DiGirolamo, C.M., and Prockop, D.J.
Rapid expansion of recycling stem cells in cultures of plastic-
adherent cells from human bone marrow. Proc Natl Acad Sci
USA97, 3213, 2000.
30. de Bruijn, J.D., van den Brink, I., Mendes, S., Dekker, R.,
Bovell, Y.P., and van Blitterswijk, C.A. Bone induction by
implants coated with cultured osteogenic bone marrow cells.
Adv Dent Res 13, 74, 1999.
Address reprint requests to:
Joost de Bruijn, Ph.D.
Department of Materials
Queen Mary University of London
Mile End Road
London E1 4NS
United Kingdom
E-mail: j.d.debruijn@gmul.ac.uk
EXPANSION OF MESENCHYMAL STEM CELLS 9
... The increase in localized densities can lead to both cell-cell contact inhibition (Fossett and Khan, 2012;Jossen et al., 2018), genome instability or epigenetic changes (e.g., DNA methylation (Redaelli et al., 2012)), and even to cell death (Sekimoto et al., 2019). Furthermore, lower seeding densities would result in a better performance during the cultivation (Bartmann et al., 2007;Both et al., 2007;Fossett and Khan, 2012). However, for a higher initial seeding density there is a tradeoff between accelerating the cultivation due to a higher cell number in the first place and inhibiting the growth due to contact inhibition and lack of nutrient for the high cell numbers (Bartmann et al., 2007;Both et al., 2007;Fossett and Khan, 2012). ...
... Furthermore, lower seeding densities would result in a better performance during the cultivation (Bartmann et al., 2007;Both et al., 2007;Fossett and Khan, 2012). However, for a higher initial seeding density there is a tradeoff between accelerating the cultivation due to a higher cell number in the first place and inhibiting the growth due to contact inhibition and lack of nutrient for the high cell numbers (Bartmann et al., 2007;Both et al., 2007;Fossett and Khan, 2012). Therefore, a high initial seeding density with a good overall dispersion is beneficial for the cultivation process. ...
Article
Current seeding processes of mesenchymal stem cells (MSC) in 2D in vitro culture systems rely on the experience and practice of experimentalists. A systematic understanding of the operation of this process is needed especially considering future scale-out and automation. This work presents a computational fluid dynamics (CFD) model-based design of seeding processes for 2D mesenchymal stem cell cultivation. The CFD-based process model enables the investigation of the cell heterogeneity at local and global levels using visual and numerical analyses. Case studies were performed to single out the impact of changing experimental design variables on the seeding heterogeneity. A significant improvement could be achieved by positioning the pipette right above the center of the plate, increasing the initial media level, and lowering the pipette angle at an initial seeding density of 5000 cells cm–2. For ideal seeding conditions, temperature differences between the cultivation media and the environment should be avoided.
... Control of the cell cycle is necessary to ensure correct cell development and function. In vitro studies demonstrate that cell passage and density influence the proliferation rate; for example, younger cells at lower density proliferate faster [158]. Another important factor that decreases cell proliferation rate is time in culture, which affects telomerase activity and leads to morphological and molecular changes in the cell. ...
Article
Full-text available
Mesenchymal stromal cell (MSC)-based advanced therapy medicinal products (ATMPs) are being tried in a vast range of clinical applications. These cells can be isolated from different donor tissues by using several methods, or they can even be derived from induced pluripotent stem cells or embryonic stem cells. However, ATMP heterogeneity may impact product identity and potency, and, consequently, clinical trial outcomes. In this review, we discuss these topics and the need to establish minimal criteria regarding the manufacturing of MSCs so that these innovative therapeutics may be better positioned to contribute to the advancement of regenerative medicine.
Article
Three-dimensional (3D) cell assemblies, such as multicellular spheroids, can be powerful biological tools to closely mimic the complexity of cell–cell and cell–matrix interactions in a native-like microenvironment. However, potential applications of large spheroids are limited by the insufficient diffusion of oxygen and nutrients through the spheroids and, thus, result in the formation of a necrotic core. To overcome this drawback, we present a new strategy based on nanoparticle-coated microparticles. In this study, microparticles function as synthetic centers to regulate the diffusion of small molecules, such as oxygen and nutrients, within human mesenchymal stem cell (hMSC) spheroids. The nanoparticle coating on the microparticle surface acts as a nutrient reservoir to release glucose locally within the spheroids. We first coated the surface of the poly(lactic-co-glycolic acid) (PLGA) microparticles with mesoporous silica nanoparticles (MSNs) based on electrostatic interactions and then formed cell-nanofunctionalized microparticle spheroids. Next, we investigated the stability of the MSN coating on the microparticles’ surface during 14 days of incubation in cell culture medium at 37 °C. Then, we evaluated the influence of MSN-coated PLGA microparticles on spheroid aggregation and cell viability. Our results showed the formation of homogeneous spheroids with good cell viability. As a proof of concept, fluorescently labeled glucose (2-NBD glucose) was loaded into the MSNs at different concentrations, and the release behavior was monitored. For cell culture studies, glucose was loaded into the MSNs coated onto the PLGA microparticles to sustain local nutrient release within the hMSC spheroids. In vitro results demonstrated that the local delivery of glucose from MSNs enhanced the cell viability in spheroids during a short-term hypoxic culture. Taken together, the newly developed nanofunctionalized microparticle-based delivery system may offer a versatile platform for local delivery of small molecules within 3D cellular assemblies and, thus, improve cell viability in spheroids.
Article
Full-text available
The treatment of fungal bone infections and infected non-unions is a huge challenge in modern trauma and orthopedics, which normally contain the local and systemic administration of anti-fungal drugs. Although frequently used, little is known about the impact of systemic and locally administered fungicides on the osteogenic regenerative capabilities of infected bone tissue, especially upon the osteogenesis of human bone marrow mesenchymal stem cells (BM-hMSCs). This study evaluates the effects of the three most common fungicides for the systemic treatment of bone infections, Voriconazole (VOR), liposomal Amphotericin B (LAMB), and Fluconazole (FLU), as well as the effects of VOR and LAMB-loaded Polymethylmethacrylate (PMMA) cement chips in different concentrations upon the osteogenic response of BM-hMSCs in vitro. Within this study, we compared the ability of BM-hMSC to differentiate into osteoblast-like cells and synthesize hydroxyapatite as assessed by radioactive 99mTechnetium-Hydroxydiphosphonate (99mTc-HDP) labeling, cell proliferation, and analyses of supernatants upon various osteogenic parameters. Our results revealed that VOR added to the cell culture medium affects the osteogenic potential of BM-hMSC negatively, while there were no detectable effects of LAMB and FLU. Moreover, we showed dose-dependent negative effects of high- and extended-dose fungicide-loaded PMMA cement due to cytotoxicity, with a higher cytotoxic potential of VOR than LAMB, while low-dose fungicide-loaded PMMA had no significant effect on the osteogenic potential of BM-hMSC in vitro.
Article
Full-text available
Tendon is a highly organized tissue that transmits forces between muscle and bone. The architecture of the extracellular matrix of tendon, predominantly from collagen type I, is important for maintaining tenocyte phenotype and function. Therefore, in repair and regeneration of damaged and diseased tendon tissue, it is crucial to restore the aligned arrangement of the collagen type I fibers of the original matrix. To this end, a novel, user‐friendly microfluidic piggyback platform is developed allowing the controlled patterned formation and alignment of collagen fibers simply on the bottom of culture dishes. Rat tenocytes cultured on the micropatterns of aligned fibrous collagen exhibit a more elongated morphology. The cells also show an increased expression of tenogenic markers at the gene and protein level compared to tenocytes cultured on tissue culture plastic or non‐fibrillar collagen coatings. Moreover, using imprinted polystyrene replicas of aligned collagen fibers, this work shows that the fibrillar structure of collagen per se affects the tenocyte morphology, whereas the biochemical nature of collagen plays a prominent role in the expression of tenogenic markers. Beyond the controlled provision of aligned collagen, the microfluidic platform can aid in developing more physiologically relevant in vitro models of tendon and its regeneration.
Article
Full-text available
Nucleic acids have clear clinical potential for gene therapy. Plasmid DNA (pDNA) was the first nucleic acid to be pursued as a therapeutic molecule. Recently, mRNA came into play as it offers improved safety and affordability. In this study, we investigated the uptake mechanisms and efficiencies of genetic material by cells. We focused on three main variables (1) the nucleic acid (pDNA, or chemically modified mRNA), (2) the delivery vector (Lipofectamine 3000 or 3DFect), and (3) human primary cells (mesenchymal stem cells, dermal fibroblasts, and osteoblasts). In addition, transfections were studied in a 3D environment using electrospun scaffolds. Cellular internalization and intracellular trafficking were assessed by using enhancers or inhibitors of endocytosis and endosomal escape. The polymeric vector TransIT-X2 was included for comparison purposes. While lipoplexes utilized several entry routes, uptake via caveolae served as the main route for gene delivery. pDNA yielded higher expression levels in fast-dividing fibroblasts, whereas, in slow-dividing osteoblasts, cmRNA was responsible for high protein production. In the case of mesenchymal stem cells, which presented an intermediate doubling time, the combination vector/nucleic acid seemed more relevant than the nucleic acid per se. In all cases, protein expression was higher when the cells were seeded on 3D scaffolds.
Article
Full-text available
Engineered living microtissues such as cellular spheroids and organoids have enormous potential for the study and regeneration of tissues and organs. Microtissues are typically engineered via self‐assembly of adherent cells into cellular spheroids, which are characterized by little to no cell–material interactions. Consequently, 3D microtissue models currently lack structural biomechanical and biochemical control over their internal microenvironment resulting in suboptimal functional performance such as limited stem cell differentiation potential. Here, this work report on stimuli‐responsive cell‐adhesive micromaterials (SCMs) that can self‐assemble with cells into 3D living composite microtissues through integrin binding, even under serum‐free conditions. It is demonstrated that SCMs homogeneously distribute within engineered microtissues and act as biomechanically and biochemically tunable designer materials that can alter the composite tissue microenvironment on demand. Specifically, cell behavior is controlled based on the size, stiffness, number ratio, and biofunctionalization of SCMs in a temporal manner via orthogonal secondary crosslinking strategies. Photo‐based mechanical tuning of SCMs reveals early onset stiffness‐controlled lineage commitment of differentiating stem cell spheroids. In contrast to conventional encapsulation of stem cell spheroids within bulk hydrogel, incorporating cell‐sized SCMs within stem cell spheroids uniquely provides biomechanical cues throughout the composite microtissues’ volume, which is demonstrated to be essential for osteogenic differentiation.
Article
Full-text available
Focal adhesions are large macromolecular assemblies through which cells are connected with the extracellular matrix so that extracellular signals can be transmitted inside cells. Some studies have focused on the effect of cell shape on the differentiation of stem cells, but little attention has been paid to focal adhesion. In the present study, mesenchymal stem cells (MSCs) and osteoblast-like MC3T3-E1 cells were seeded onto micropatterned substrates on which circular adhesive islands with different spacing and area were created for focal adhesion. Results showed that the patterns of focal adhesion changed cell morphology but did not affect cell survival. For MSCs cultured for 3 days, patterns with small circles and large spacing promoted osteogenesis. For MSCs cultured for 7 days, patterns with large circles and spacing enhanced osteogenesis. For MC3T3-E1 cells, the patterns of focal adhesion had no effect on cell differentiation after 3 days of culture, but patterns with small circles and spacing improved osteogenic differentiation after 7 days. Moreover, the assembly of F-actin, phosphorylation of myosin, and nuclear translocation of yes-associated proteins (YAP) were consistent with the expression of differentiation markers, indicating that the pattern of focal adhesion may affect the osteogenesis of MSCs and osteoblasts through changes in cytoskeletal tension and nuclear localisation of YAP.
Article
Full-text available
In biomaterials R&D, conventional monolayer cell culture on flat/planar material samples, such as films, is still commonly employed at early stages of the assessment of interactions of cells with candidate materials considered for a biomedical application. In this feasibility study, an approach for the assessment of 3D cell–material interactions through dispersed coaggregation of microparticles from biomaterials into tissue spheroids is presented. Biomaterial microparticles can be created comparatively quickly and easily, allow the miniaturization of the assessment platform, and enable an unhindered remodeling of the dynamic cell–biomaterial system at any time. The aggregation of the microsized biomaterials and the cells is supported by low‐attachment round‐bottom microwells from thin polymer films arranged in densely packed arrays. The study is conducted by the example of MG63 osteoblast‐like and human mesenchymal stem/stromal cells, and a small library of model microbiomaterials related to bone repair and regeneration. For the proof of concept, example interactions including cell adhesion to the material, the hybrid spheroids’ morphology, size, and shape, material‐associated cell death, cell metabolic activity, cell proliferation, and (osteogenic) differentiation are investigated. The cells in the spheroids are shown to respond to differences in the microbiomaterials’ properties, their amounts, and the duration of interaction with them.
Article
Full-text available
Advanced therapy medicinal products (ATMPs) offer new prospects to improve the treatment of conditions with unmet medical needs. Kidney diseases are a current major health concern with an increasing global prevalence. Chronic renal failure appears after many years of impairment, which opens a temporary window to apply novel therapeutic approaches to delay or halt disease progression. The immunomodulatory, anti-inflammatory, and pro-regenerative properties of mesenchymal stromal cells (MSCs) have sparked interest for their use in cell-based regenerative therapies. Currently, several early-phase clinical trials have been completed and many are ongoing to explore MSC safety and efficacy in a wide range of nephropathies. However, one of the current roadblocks to the clinical translation of MSC therapies relates to the lack of standardization and harmonization of MSC manufacturing protocols, which currently hinders inter-study comparability. Studies have shown that cell culture processing variables can have significant effects on MSC phenotype and functionality, and these are highly variable across laboratories. In addition, heterogeneity within MSC populations is another obstacle. Furthermore, MSCs may be isolated from several sources which adds another variable to the comparative assessment of outcomes. There is now a growing body of literature highlighting unique and distinctive properties of MSCs according to the tissue origin, and that characteristics such as donor, age, sex and underlying medical conditions may alter the therapeutic effect of MSCs. These variables must be taken into consideration when developing a cell therapy product. Having an optimal scale-up strategy for MSC manufacturing is critical for ensuring product quality while minimizing costs and time of production, as well as avoiding potential risks. Ideally, optimal scale-up strategies must be carefully considered and identified during the early stages of development, as making changes later in the bioprocess workflow will require re-optimization and validation, which may have a significant long-term impact on the cost of the therapy. This article provides a summary of important cell culture processing variables to consider in the scale-up of MSC manufacturing as well as giving a comprehensive review of tissue of origin-specific biological characteristics of MSCs and their use in current clinical trials in a range of renal pathologies.
Article
Human bone marrow contains a population of cells capable of differentiating along multiple mesenchymal cell lineages. Recently, techniques for the purification and culture-expansion of these human marrow-derived Mesenchymal Stem Cells (MSCs) have been developed. The goals of the current study were to establish a reproducible system for the in vitro osteogenic differentiation of human MSCs, and to characterize the effect of changes in the microenvironment upon the process. MSCs derived from 2nd or 3rd passage were cultured for 16 days in various base media containing 1 to 1000 nM dexamethasone (Dex), 0.01 to 4 mM L-ascorbic acid-2-phosphate (AsAP) or 0.25 mM ascorbic acid, and 1 to 10 mM beta-glycerophosphate (beta GP). Optimal osteogenic differentiation, as determined by osteoblastic morphology, expression of alkaline phosphatase (APase), reactivity with anti-osteogenic cell surface monoclonal antibodies, modulation of osteocalcin mRNA production, and the formation of a mineralized extracellular matrix containing hydroxyapatite was achieved with DMEM base medium plus 100 nM Dex, 0.05 mM AsAP, and 10 mM beta GP. The formation of a continuously interconnected network of APase-positive cells and mineralized matrix supports the characterization of this progenitor population as homogeneous. While higher initial seeding densities did not affect cell number of APase activity, significantly more mineral was deposited in these cultures, suggesting that events which occur early in the differentiation process are linked to end-stage phenotypic expression. Furthermore, cultures allowed to concentrate their soluble products in the media produced more mineralized matrix, thereby implying a role for autocrine or paracrine factors synthesized by human MSCs undergoing osteoblastic lineage progression. This culture system is responsive to subtle manipulations including the basal nutrient medium, dose of physiologic supplements, cell seeding density, and volume of tissue culture medium. Cultured human MSCs provide a useful model for evaluating the multiple factors responsible for the step-wise progression of cells from undifferentiated precursors to secretory osteoblasts, and eventually terminally differentiated osteocytes.
Article
A simple method for preparing undecalcified thin sections of bone with implants has been developed. After exposing a surface of bone and implant in a plastic block by sawing thick sections, the surface is stained prior to making a thin section. A glass coverslip is affixed with a thin layer of cement to the stained surface to stabilize the tissue and implant during sectioning. A mixture of glycerine and water is used as a coolant and lubricant. The orientation in situ is preserved allowing demonstration of bone architecture and cells, and the tissue-implant interface. © 1988 Informa UK Ltd All rights reserved: reproduction in whole or part not permitted.
Article
Investigation of osteoblast dysfunction in osteoporosis has been hampered by a poor understanding of normal early osteoblast differentiation, due to a relative lack of markers for the earliest cells in the lineage. Attempts to identify such markers have used cultures of animal or immortalized human cells, of uncertain relevance to human biology, or heterogeneous cultures in which genetic variability precludes the isolation of stage-specific genotypic markers. Primary in vitro generation of clonal populations of human bone marrow stromal cells was used in order to overcome these problems. Fibroblast-like stromal cells were isolated from human sternal bone marrow. They showed differentiation to an osteoblastic phenotype when stimulated with dexamethasone (10−7 m) and fluorescence activated cell analysis demonstrated immunopositivity for STRO-1 (an antibody that recognizes osteoprogenitor stem cells of the colony-forming unit-fibroblastic) in from 8 to 40 per cent of the cells, dependent on time post-harvest. Cells positive for STRO-1 were immunoselected using magnetic activated cell sorting and seeded at low density (10 cells/cm2 ) to produce clones. Each clone was subpassaged, osteoblastic differentiation stimulated with dexamethasone, and mRNA-extracted at time points post-stimulation (0h and 1–14 days). A novel poly (A) reverse transcriptase-polymerase chain reaction (RT-PCR) was used to amplify cDNA representative of all transcripts expressed at each time point. Differential gene expression within the amplified cDNA was assessed using 3′ end cDNA probes to osteocalcin, osteopontin, and collagen type I (positive), demonstrating the acquisition of an osteoblastic phenotype. Time-specific gene products for early osteoblast differentiation have been generated from primary human cultures, utilizing very low density seeding and poly (A) RT-PCR. These products overcome the problems associated with animal, immortalized or heterogeneous culture and can be used to study normal and altered early osteoblast differentiation, indicating the possibility of using the same system to study other disease states. Copyright © 1999 John Wiley & Sons, Ltd.
Article
Human marrow stromal cells (MSCs) were isolated from posterior illiac crest marrow aspirates obtained from 17 healthy donors, ages 19–45 years, with no apparent physical disability. First passage hMSCs exhibited growth rates in vitro that varied up to 12-fold between donors. No correlation between growth rate and the age or gender of the donor was evident (P ≤ 0.05). When hMSCs were cultured without passage for eight days (subconfluent cultures) or 22 days (confluent cultures) in the absence of any osteogenic agonists, levels of alkaline phosphatase enzyme activity varied 40-fold and 10-fold, respectively, between donors. When exposed to osteo-inductive media, donor populations also showed dramatic differences in levels of bone-specific gene induction. Collectively, these data demonstrate that hMSC cultures are composed of a heterogeneous mixture of cells at various stages of differentiation and with distinct osteogenic potentials. Differences in both growth rate and ALP activity were evident in hMSC cultures established from multiple aspirates obtained over a six month period from the same donors. Therefore, it appears that cellular heterogeneity produced by the method of harvest is propagated within and among different donor populations during culture expansion in vitro. J. Cell. Biochem. 75:424–436, 1999. © 1999 Wiley-Liss, Inc.
Article
Using the in vitro colony assay, clonogenic fibroblast precursor cells (CFU-F) were detected in the bone marrow, spleen and thymus from adult mice. The survival curve for CFU-F of mouse bone marrow irradiated in vitro has a D0 of 220 r. Regeneration of bone marrow CFU-F after whole-body irradiation with 150 r is characterized by a marked secondary loss and post-irradiation lag and dip, lasting 6 days, followed by return to normal values by about the 25th day. This pattern of post-radiation recovery of CFU-F is similar to that of the CFU-s. In addition, during the first 6 hours following irradiation the number of CFU-F increased approximately twofold.
Article
Studies using animal tissue suggest that bone marrow contains cells with the potential to differentiate into cartilage and bone. We report the extension of these studies to include human marrow. Bone marrow from male and female donors of various ages was obtained either from the femoral head or as aspirates from the iliac crest, and introduced into culture. Culture-adherent cells were expanded, subcultured, and then tested for bone and cartilage differentiation potential utilizing two different in vivo assays in nude mice. One assay involved subcutaneous implantation of porous calcium phosphate ceramics loaded with cultured, marrow-derived, mesenchymal cells; the other involved peritoneal implantation of diffusion chambers, also inoculated with cultured, marrow-derived, mesenchymal cells. Histological evaluation showed bone formation in ceramics implanted with cultured, marrow-derived, mesenchymal cells originating from both the femoral head and the iliac crest. Immunocytochemical analysis indicates that the bone is derived from the implanted human cells and not from the cells of the rodent host. No cartilage was observed in any of these ceramic grafts. In contrast, aliquots from the same preparations of cultured, marrow-derived, mesenchymal cells failed to form bone or cartilage in diffusion chambers. These data suggest that human marrow contains cells with osteogenic potential, which can be enriched and expanded in culture. Our findings also suggest that subcutaneous implantation of these cells in porous calcium phosphate ceramics may be a more sensitive in vivo assay than diffusion chambers for measuring their osteogenic lineage potential.
Article
A simple method for preparing undecalcified thin sections of bone with implants has been developed. After exposing a surface of bone and implant in a plastic block by sawing thick sections, the surface is stained prior to making a thin section. A glass coverslip is affixed with a thin layer of cement to the stained surface to stabilize the tissue and implant during sectioning. A mixture of glycerine and water is used as a coolant and lubricant. The orientation in situ is preserved allowing demonstration of bone architecture and cells, and the tissue-implant interface.
Article
Evidence is discussed for the hypothesis that there are stromal stem cells present in the soft connective tissues associated with marrow and bone surfaces that are able to give rise to a number of different cell lines including the osteogenic line. Fibroblastic colonies, each derived from a single colony-forming unit fibroblastic (CFU-F), are formed when marrow cells are cultured in vitro. In vivo assays of CFU-F have demonstrated that some CFU-F have a high ability for self renewal and multipotentiality whereas some have more limited potential. In vitro studies also support the hypothesis and have shown that CFU-F are a heterogeneous population of stem and progenitor cells and that their differentiation in vitro can be modified at the colony level. Factors added to the medium can activate osteogenesis in a range of multipotential and more committed precursors. Different stromal cell lines can be promoted under different culture conditions. The number and hierarchy of cell lines belonging to the stromal fibroblastic system are not yet fully elucidated and more specific markers for the different lines are required before a better understanding can be achieved.
Article
Cells from fetal or neonatal skeleton can synthesize bone-like tissue in vitro. In contrast, formation of bone-like tissue in vitro by cells derived from adult animals has rarely been reported and has not been achieved using cells from bone marrow. We have explored development of bone-like tissue in vitro by bone marrow stromal cells. Marrow stromal cells obtained from 40-43-day-old Wistar rats were grown in primary culture for 7 days and then subcultured for 20-30 days. Cells were cultured in either alpha-minimal essential medium containing 15% fetal bovine serum, antibiotics, and 50 micrograms/ml ascorbic acid, or the above medium supplemented with either 10 mM Na-beta-glycerophosphate, 10(-8) M dexamethasone, or a combination of both. Cultures were examined using phase-contrast microscopy, undemineralized and demineralized tissue histology, histochemistry (for alkaline phosphatase activity), immunohistochemistry (for collagen type, osteonectin, and bone Gla-protein), scanning and transmission electron microscopy, energy dispersive X-ray microanalysis, and X-ray diffraction. Collagenous, mineralized nodules exhibiting morphological and ultrastructural characteristics similar to bone were formed in the cultures, but only in the presence of both beta-glycerophosphate and dexamethasone. Cells associated with the nodules exhibited alkaline phosphatase activity. The matrix of the nodules was composed predominantly of type-I collagen and both osteonectin and Gla-protein were present. X-ray microanalysis showed the presence of Ca and P, and X-ray diffraction indicated the mineral to be hydroxyapatite. The nodules were also examined for bone morphogenetic protein-like activity. Paired diffusion chambers containing partly demineralized nodules and fetal muscle were implanted intraperitonealy in rats. Induction of cartilage in relation to muscle was observed histologically after 40 days in the chambers. This finding provided further support for the bone-like nature of the nodules. The observations show that bone-like tissue can be synthesized in vitro by cells cultured from young-adult bone marrow, provided that the medium contains both beta-glycerophosphate and, particularly, dexamethasone.
Article
Fibroblast colonies (clones) were obtained by explantation of bone marrow single-cell suspensions and were used to establish multicolony and single-colony derived fibroblast cultures by successive passaging of either pooled or individual colonies. When transplanted in diffusion chambers after 20-30 cell doublings in vitro, the descendants of fibroblast colony-forming cells (FCFC), whether grown from single or pooled colonies, retained the ability for bone and cartilage formation. The content of osteogenic precursors in the cultured progeny significantly outnumbered the initiating FCFC. Thus the high proliferative potential of bone marrow FCFC and their ability to serve as common precursors of bone and cartilage-forming cells makes them probable candidates for the role of osteogenic stem cells.