Vous êtes sur la page 1sur 10

Biotechnol. Appl. Biochem. (2010) 55, 199208 (Printed in Great Britain) doi:10.

1042/BA20090356 199

Isolation of a mouse bone marrow population enriched in stem


and progenitor cells by centrifugation on a Percoll gradient

Ana-Maria Rosca and Alexandrina Burlacu1

Laboratory for Adult and Embryonic Stem Cell Biology, Institute of Cellular Biology and Pathology Nicolae Simionescu, Hasdeu
Street, Bucharest 050568, Romania

Given the complex composition of bone marrow, a [3]. However, stem/progenitor cells represent rare events
cell separation technique that results in populations among bone marrow nucleated cells [4,5]. They exist in a
enriched in progenitor cells is required for cellular dif- highly organized microenvironment composed of stromal
ferentiation and transplantation studies. In the present cells (osteoblasts, adipocytes, endothelial cells and vascular
study, we designed a method that allows for the pericytes), an extracellular matrix rich in fibronectin,
isolation of a progenitor-enriched population of bone collagens and proteoglycans [6] and a large amount of
marrow by exploiting the physical properties of these haematopoietic cells.
cells. Bone marrow aspirate was separated on a discon- Given the complex composition of bone marrow, a
tinuous Percoll gradient (ranging from 1.050 to 1.083 cell separation technique that results in subpopulations
g/cm3 ) that resulted in the recovery of six cell fractions. enriched in stem/progenitor cells is required for cellular
The fractions were characterized by FACS and RTPCR differentiation and transplantation studies. At present,
(reverse transcriptionPCR) analyses and evaluated for density gradient centrifugation using Histopaque is employed
their capacity to differentiate into haematopoietic and by many clinical investigators to separate progenitor cells
mesenchymal cells. Fraction IV, including cells with from blood; however, it provides a pool of mononuclear cells
a density of 1.0701.076 g/ml, contained 11.68% of containing all cells with a density of less than 1.077 g/ml, but
total bone marrow cells and was enriched in c-kit+ no substantial improvement in progenitors. Other general
and Sca-1+ (stem cell antigen-1) progenitor cells as methods to isolate progenitor cells from the mononuclear
compared with total bone marrow. This fraction cell population include immunological methods [7], which
demonstrated an increase in clonogenic capacity under are time-consuming and expensive because of the use of
specific conditions as well as a potential to generate a monoclonal antibodies and advanced technology.
mesenchymal stem cell culture in a shorter period than In the present study, we aimed to find out whether
that using bone marrow aspirate. Furthermore, this a bone marrow subpopulation enriched in stem/progenitor
fraction lacked differentiated cell types and contained cells can be obtained by exploiting the physical properties
cells positive for endothelial markers, which further of these cells. For this purpose, we designed a method to
increases its value in cellular transplant. In conclusion, fractionate bone marrow cells, aiming in particular for cells
a bone marrow subpopulation that is enriched in with a density of less than 1.077 g/ml. The bone marrow
progenitor cells and may be valuable in cellular suspension was layered on a discontinuous Percoll gradient,
transplant therapy can be isolated by exploiting the which allowed the fractionation of cells with a density of
physical properties of these cells. less than 1.077 g/ml into five subpopulations. Cells with
a density higher than 1.077 g/ml remained at the bottom
of the gradient in a distinct fraction. We showed that
a subpopulation of bone marrow, which was enriched in
Introduction small, c-kit+ and Sca-1+ (stem cell antigen-1) progenitor

Adult bone marrow is still recognized as the main source


of stem/progenitor cells throughout the life of an adult. Key words: bone marrow, c-kit, haematopoietic stem cell (HSC),
This tissue houses two stem cell populations with distinct mesenchymal stem cell (MSC), Percoll, stem cell antigen-1 (Sca-1).
progenies that are valuable in cellular transplant: HSCs Abbreviations used: CFU, colony forming units; DMEM, Dulbeccos modified
Eagles medium; FBS, fetal bovine serum; HSC, haematopoietic stem cell;
(haematopoietic stem cells) and MSCs (mesenchymal stem MSC, mesenchymal stem cell; PBS-FBS, PBS containing 2% FBS; PECAM,
cells) [1]. HSCs can give rise to all blood cells, whereas platelet endothelial cell adhesion molecule; RTPCR, reverse
MSCs have been shown to differentiate in vitro, not only transcriptionPCR; Sca-1, stem cell antigen-1; SSC, side scatter; VEGFR2,
vascular endothelial growth factor receptor 2.
into mesenchymal lineages, such as osteoblasts, adipocytes 1
To whom correspondence should be addressed (email
and chondrocytes [1,2], but also into neurons and muscle sanda.burlacu@icbp.ro).


C 2010 Portland Press Limited
200 A.-M. Rosca and A. Burlacu

cells, could be isolated by Percoll gradient centrifugation. was used at 10 g/ml. Incubation with the primary antibody
This subpopulation, containing cells with a density varying was followed by quenching in 3% H2 O2 and incubation
between 1.067 and 1.070 g/ml, had an increased capacity with horseradish peroxidase-conjugated anti-mouse IgG.
to generate haematopoietic cells under specific conditions Ultimately, the proteins were visualized by the DAB
and, moreover, produced MSC cultures that were free of (diaminobenzidine) reaction and examined under a Nikon
haematopoietic cells in a shorter period compared with microscope.
those from bone marrow aspirate.
Flow cytometry assay
Materials and methods FACS was performed using a MoFlo Cell Sorter. Cells
from fractions IVI and bone marrow aspirate were
Isolation of bone marrow cells incubated with a phycoerythrin-labelled anti-c-kit or anti-
Adult RAP mice (8 weeks old) were killed by cervical Sca-1 antibody (R&D Systems) for 1 h at 4 C, washed in
dislocation in accordance with the rules of the Institutional PBS-FBS and analysed. In order to exclude dead cells from
Ethical Board for Experimental Procedures. The tibiae and the analysis, 1 g/ml propidium iodide was added just before
femurs were harvested, and the medullar channels were the analysis. At least 20000 events were considered for each
flushed with 5 ml PBS-FBS [PBS containing 2% FBS (fetal sample. Results were analysed using Summit v4.3 software
bovine serum)]. The single cell suspension obtained by (Cytomation, Inc).
mechanical dissociation (passing the cells through needles
of decreasing gauges, i.e. 20, 22 and 25 gauge) was then RTPCR (reverse transcriptionPCR)
centrifuged at 400 g for 5 min at 20 C and resuspended Total RNA was extracted using the RNeasy Micro kit (Qia-
in 1 ml of DMEM (Dulbeccos modified Eagles medium; gen), and cDNA was synthesized from 0.2 g of total RNA
Invitrogen). This volume of suspension was layered on the employing MMLV Reverse transcriptase (Invitrogen). PCR
Percoll gradient or analysed by flow cytometry. was performed using optimized amplification conditions, and
the products were visualized on a 1.5% agarose gel with
Separation on the Percoll gradient ethidium bromide staining. For each gene, DNA primers
An isotonic Percoll solution was made by mixing Percoll (25 were derived from different exons whenever possible, and
mOs/kg, 1.130 g/ml; SigmaAldrich) and 10 concentrated cDNAs were treated with DNase I to ensure that the PCR
DMEM at a 9:1 volume ratio. Densities of 1.050, 1.057, product represented the specific messenger RNA species
1.067, 1.070, 1.076 and 1.083 g/ml were prepared by diluting and not genomic DNA. The primer sequences are available
the isotonic Percoll solution in DMEM at concentrations upon request.
of 35, 40, 45, 50, 55 and 60% respectively. A six-layered
Percoll gradient was obtained by layering 2 ml of each Colony-forming unit assay
Percoll solution. Finally, the cell suspension obtained as The functionality of the haematopoietic stem/progenitor
described above was placed on the top of the gradient and cells within each fraction was estimated in HSC-CFU
centrifuged at 1500 g for 25 min at 4 C. Five fractions of complete with Epo medium (Miltenyi Biotec). This medium
2 ml each were collected after centrifugation and designated allows the enumeration of haematopoietic stem and
IV (see Figure 2A). The lower part of the gradient (3 progenitor cells characterized as CFU (colony forming units)
ml), which contained the cells with a density higher than and has been reported to support the growth of granulocyte
1.076g/ml and included the erythrocytes, was collected as (CFU-G), macrophage (CFU-M), granulocyte/macrophage
fraction VI. The cells in each fraction were washed three (CFU-GM) and erythroid [BFU-E (erythroid burst-forming
times in PBS-FBS and then either analysed or cultivated units) and CFU-E] colonies, as well as mixed colonies (CFU-
in DMEM supplemented with 10% MSC-Qualified FBS GEMM) [8]. Cells were cultivated at 1500 cells/cm2 in
(Invitrogen). duplicates and clusters of cells were scored after 7 days.

In situ detection of progenitor cells Adipogenic and osteogenic differentiation


Tibiae were fixed in formalin and then decalcified in Cells from each fraction were seeded into two wells of a
5% trichloroacetic acid, dehydrated in a graded series 24-well plate. After 1 week in culture, the cells were
of alcohols and embedded in paraffin. Longitudinal incubated for an additional week in either adipogenic
sections (5 m) on poly-L-lysine-coated glass slides were [DMEM with 10% FBS, 106 M dexamethasone, 100 M
deparaffinized and microwaved in citrate buffer for antigen indomethancin and 1% ITS (insulin-transferrin-sodium
retrieval. Anti-mouse c-kit or Sca-1 antibody (R&D Systems) selenite supplement)] [9] or osteogenic (DMEM with 10%

C 2010 Portland Press Limited
Isolation of a progenitor-enriched population of bone marrow 201

Figure 1 Quantification and localization of c-kit+ and Sca-1+ cells in bone marrow

(A) Flow-cytometry quantification of c-kit+ and Sca-1+ cells in bone marrow. C-kit was identified on 11.8 +
2.5% of the bone marrow cells with varying granularity,
whereas Sca-1+ cells represented 9.8 + 2.8% of bone marrow cells and were almost exclusively characterized by low granularity. Diagrams are representatives of
four independent experiments, whose mean values are illustrated in the adjoined table. (B) Immunohistochemistry for in situ localization of c-kit+ (b and c) and
Sca-1+ (d, e and f) cells within bone marrow. Staining was completely absent in tissue sections in which the primary antibody was omitted (a). One can notice the
epiphysis as preferential site for Sca-1+ cells (df), and the diaphysis for c-kit+ cells (b and c).

FBS, 107 M dexamethasone, 10 mM -glycerophosphate Results


and 0.3 mM ascorbic acid) [6] differentiation medium.
All chemicals were from Sigma, unless otherwise stated. Localization and quantification of c-kit- and
Lipid droplets were stained with Oil Red (Sigma), and Sca-1-positive cells in mouse bone marrow
lipid accumulation was quantified as previously described Quantification of progenitor cells from the bone marrow
[10] using the TECAN iGenios spectrofluorimeter. Calcium aspirate was determined on the basis of c-kit and Sca-1
deposits were stained by the von Kossa method [6]. surface marker expression. Flow cytometry results revealed
that 11.8% of total bone marrow cells were c-kit+ cells
and 9.8% were Sca-1+ cells (Figure 1A). To localize their
Statistics preferential site within bone marrow, longitudinal sections
All quantitative results were expressed as the of decalcified tibiae were analysed by immunohistochemistry
means + S.E.M. from at least three experiments. Statistical (Figure 1B). c-kit+ cells were predominantly localized in
analysis was performed using one-way ANOVA. A p-value the diaphyseal region (Figure 1B, b and c), whereas Sca-
less than 0.05 was considered significant. 1+ cells were detected within the epiphyseal region of


C 2010 Portland Press Limited
202 A.-M. Rosca and A. Burlacu

Figure 2 Separation of bone marrow cells into six fractions by centrifugation on a discontinuous Percoll gradient

(A) Schematic illustration of the discontinuous Percoll gradient used for fractionation and the percentage of cells segregated in each fraction after centrifugation.
(B) Dot-plot histograms resulting from FACS analysis illustrating the heterogeneity of cells before separation and the morphology of cells in each fraction.

the bone (Figure 1B, df). These results indicate that harvested from one mouse). The segregation of cells within
colonies of progenitor cells can be found in both the each fraction is represented in Figure 2(A) as a percentage
epiphysis and diaphysis, but differ in their expression of cell of the total amount of cells recovered after separation.
surface markers. The segregation of the two populations The counting revealed that 0.58%, 0.62%, 1.73%, 11.68%,
of progenitor cells in different sites within bone marrow 66.32% and 19.08% of cells were segregated in fractions
ruled out the existence of c-kit+ /Sca-1+ double-positive IVI respectively, meaning 2 105 cells in fraction I, 2.2 105
cells in this tissue, indicating that the large majority of in fraction II, 4.2 105 in fraction III, 4.3 106 in fraction
progenitor cells were either Sca-1 or c-kit single-positive IV, 23.7 106 in fraction V, and 6.8 106 in fraction VI.
cells. Our results corroborate other studies that revealed Erythrocytes residing mainly in fraction VI were excluded
very low percentages of Sca-1+ /c-kit+ double-positive cells from the counting. Thus the majority of cells (66.32%)
within bone marrow [11,12]. segregated in fraction V, corresponding to the density
range 1.0701.076 g/ml, whereas fraction IV contained
only a minor population of cells (11.68%). As expected,
Separation of bone marrow cells on a Percoll
flow cytometry analysis of each subpopulation revealed an
gradient
increase in cell granularity [FSC (forward scatter)/SSC] with
Because c-kit+ and Sca-1+ cells have a low level of granularity
increasing Percoll density (Figure 2B).
[as revealed by low SSC (side scatter), see Figure 1A], we
considered the possibility of exploiting physical properties
in order to separate these cells. Centrifugation of cells on Quantification of the progenitor cells within
a discontinuous Percoll gradient (ranging from 1.050 to fractions
1.083 g/cm3 ) resulted in the recovery of six cell fractions Cells within each fraction were analysed for the presence of
(IVI), corresponding to the six densities of the gradient. c-kit+ and Sca-1+ progenitor cell markers by flow cytometry.
The total number of cells recovered in the six fractions As shown in Figure 3(A), the distributions of both c-kit+ and
was 35 106 + 1.4 10 cells, meaning a recovery rate
6
Sca-1+ progenitor cells were confined to fractions IIIV and
+
of 70% 2.8% (considering an average of 50 106 cells were not present in fractions I, II and VI. The quantification

C 2010 Portland Press Limited
Isolation of a progenitor-enriched population of bone marrow 203

Figure 3 Depiction of c-kit+ and Sca-1+ cells within fractions

(A) Flow cytometry diagrams revealing the percentage of c-kit+ and Sca-1+ cells within each fraction obtained after Percoll gradient separation of bone marrow
aspirate. The progenitor cells are mainly found in fractions III, IV and V. The diagram is representative of three experiments, and the average percentages are given
in the bar charts below. (B) The overall representation of flow cytometry data showing the enrichment factor for c-kit+ and Sca-1+ cells in the six fractions relative
to bone marrow aspirate (unfractionated cells). The values denote the average of three independent experiments after extraction of the unspecific binding values.
Note that fractions III and IV show the greatest enrichment for progenitor cells, illustrating 2.7- and 2.4-fold for c-kit+ cells and 1.9- and 2.2-fold for Sca-1+ cells, in
fraction III and IV respectively.

of these cells is given in Figure 3(A) as purity of each fraction aspirate. Analysis of fraction V, which contained the majority
and in Figure 3(B) as enrichment factor for c-kit+ cells (left) of the cells (66.32% of the total bone marrow cells; see
and Sca-1+ cells (right) as compared with bone marrow Figure 2A), revealed no significant increase in the percentage


C 2010 Portland Press Limited
204 A.-M. Rosca and A. Burlacu

Figure 4 Evaluation of the clonogenic potential of the haematopoietic cells within the fractions

(A) Evaluation of differentiation potential within each fraction using the CFU assay. The diagram illustrates the average value of three independent experiments.
Note that the highest capacity of differentiation resides in fractions III and IV (*P< 0.05). A representative field of fraction IV was photographed and displayed
above the diagram. (B) RTPCR illustrating the presence of representative cell types within each fraction immediately after separation (left panel) and after two
weeks in culture (right panel). As a control, c-kit+ and Sca-1+ cell populations isolated by the FACS method are given in the middle panel. (C) Lower panel:
spectrophotometric quantification of lipid accumulation in the six fractions after culturing the cells in adipogenic medium. The upper panel depicts a representative
picture of Oil Red staining in fractions III and IV. Right, light microscopy of von Kossa staining in the six fractions after cultivation in osteogenic medium. Note than
fractions III and IV, but not fraction V (that contained the majority of the cells), were able to generate osteoblasts in appropriate conditions.

of progenitor cells. Fraction V contained 14 + 1.6% c-kit


+
increase in c-kit+ cells and a 2.17 + 0.3-fold increase
+ +
and 8 4.6% Sca-1 cells as compared with 11.8 2.5% + in Sca-1+ cells in fraction IV as compared with the
c-kit+ and 9.8 + +
2.8% Sca-1 cells in the total bone marrow unfractionated bone marrow cell population. Thus fraction
cell population. In contrast, fractions III and IV contained IV contained 4.3 106 cells, of which approx. 2 106 cells
important enrichments for progenitor cells (either c-kit+ or were progenitors.
Sca-1+ ). Despite the enrichment factor of progenitor cells in To evaluate the clonogenic potential of the haemato-
fraction III (2.74 +
0.1 for c-kit cells and 1.9 +
+
0.5 for Sca-1
+
poietic cells within fraction IV, the colony-forming assay
cells), the total amount of progenitor cells in fraction III was was performed on the cell fractions. To this purpose, 1.2
inconsistent due to the low number of cells segregated in 104 cells from each fraction were plated in methylcellulose
this fraction (1.73%). Thus fraction III contained 1.8 105 medium, and the cell clusters generated were scored after
progenitor cells out of 4.2 105 cells recovered in this 7 days in culture. As illustrated in Figure 4(A, upper panel),
fraction. the colonies varied in size, probably reflecting the differences
In contrast, 23 + 4.8% of cells in fraction IV were in the proliferation rate and/or differentiation ability of the
c-kit+ and an additional 22 + +
2.3% were Sca-1 as compared various progenitor cells within the fractions. Nevertheless,
with the unfractionated population (11.8% c-kit+ cells and fractions III and IV generated the greatest number of CFU
9.8% Sca-1+ cells). These results suggested a 2.36 + 0.7-fold (Figure 4A). These fractions generated approx. 8.4 + 2.3 and

C 2010 Portland Press Limited
Isolation of a progenitor-enriched population of bone marrow 205

7.8 + 3
0.2 CFU per 10 cells, which was 2-fold higher than during cultivation. Altogether, our results demonstrate that
the major fraction, V (3.9 + 3
1.9 CFU per 10 cells). This fraction IV generated a cell culture that was depleted in
result indicates enrichment in haematopoietic progenitors haematopoietic cells, negative for osteoblast, chondrocyte
in fractions III and IV. Conversely, fractions I, II and VI and endothelial markers, yet possessed a multipotent
generated fewer CFU, as could be predicted from the capacity. These CD45 /c-kit /Sca-1+ /CD31 /Flk-1 cells are
decreased number of c-kit+ cells and Sca-1+ cells scored similar to bone marrow MSCs, which can be obtained
in those fractions. from bone marrow aspirate after several weeks in culture
[13,14].
In conclusion, a Percoll gradient can be used to
Expression of differentiated cell markers in the obtain a cell fraction that is enriched in progenitors and
fractions can generate an MSC culture after a short period of
To better characterize the content of fraction IV, the cultivation. This fraction is enriched in both haematopoietic
distribution of the main differentiated cell types that are and mesenchymal progenitor cells (c-kit+ and Sca-1+ cells),
normally present within bone marrow was evaluated in each whereas it is deprived of other differentiated cell types, and
fraction by RTPCR and the results were compared with also contains cells that are positive for endothelial markers,
c-kit+ cells and Sca-1+ cells purified using FACS. As which further increases its value in cellular transplant studies
illustrated in Figure 4(B, left panel), besides being enriched where angiogenesis is required.
in the expression of c-kit and Sca-1, fractions III and IV
showed the highest expression level of the endothelial
markers VEGFR2 (vascular endothelial growth factor Discussion
receptor 2) and PECAM (platelet endothelial cell adhesion
molecule) of all six fractions. Furthermore, fraction IV The existence of stem/progenitor cells within the hetero-
demonstrated a low level expression of differentiation geneous population of bone marrow is well documented
markers of other cell types, such as adipocytes (fabp4), [15]. Haematopoietic stem cells are the most primitive
osteoblasts (osteocalcin) and chondrocytes (collagen), cells that retain multilineage haematopoietic differentiation
which segregated mainly within fractions IIII. As expected, potential. In adult bone marrow, most haematopoietic
the FACS method generated more purified cell populations progenitors express c-kit (CD117), a member of a family
that were devoid of other cell types. of cell-surface receptors with tyrosine kinase activity
After 2 weeks in culture, adherent cells were obtai- that plays an important role in the regulation of the
ned from all fractions (results not shown). Specifically, early stages of haematopoietic development [16]. c-kit is
cells from fraction IV generated a culture of cells with a expressed in both long-term repopulating HSCs and more
fibroblast-like morphology that were distinct from those differentiated progenitor cells and is used as a valuable
derived from fractions III and V, mainly by lacking CD45 marker for the identification of these cells [17]. Although
mRNA expression (Figure 4B, right panel). Furthermore, c-kit is an established marker for HSCs, the expression
adherent cells in fraction IV expressed Sca-1 and were of Sca-1, a member of the Ly6 family, in these cells is
not contaminated with haematopoietic cells (as revealed by controversial. To date, Sca-1 expression has been identified
the absence of CD45 and c-kit mRNA) or differentiated in putative stem/progenitor cell populations within various
cells, such as endothelial cells (PECAM/CD31 and tissues and organs including bone marrow, kidney, liver,
VEGFR2/Flk-1), osteoblasts (osteocalcin) or chondrocytes lung, pancreas and muscle, as well as the aorta-gonad-
(collagen II). mesonephros, yolk sack and other regions of the embryo
To evaluate which cell fractions could generate [18]. However, whether these Sca-1+ populations are tissue-
MSC cultures, the capacity of the cells to differentiate specific precursor/stem cells or represent haematopoietic,
into adipocytes and osteoblasts was evaluated for each mesenchymal or endothelial precursor/stem cells associated
cell fraction after 1 week of cultivation. Although all with these tissues is not known in all cases [18].
fractions were able to generate adipocytes under the We show in the present paper that c-kit+ and Sca-1+
appropriate conditions, spectrophotometric quantification cells segregated within bone marrow with respect to
of lipid accumulation (Figure 4C, lower panel) suggested location, thus indicating that the large majority of progenitor
a greater capacity for fractions IIIV as compared with cells were either Sca-1 or c-kit single-positive cells. This
the other fractions. Nevertheless, the capacity to generate was also strengthened by the RTPCR analysis on FACS-
osteoblasts was limited to cells within fractions III separated c-kit+ and Sca-1+ populations that revealed the
and IV (Figure 4C, right panel), and fraction V lacked absence of c-kit mRNA in a Sca-1+ population, even if
differentiated cells. These results suggest that fractions III some c-kit+ cells do express Sca-1 at the mRNA level
and IV, but not fraction V, generated multipotent cells (Figure 4B). In accordance with these results, very low

C 2010 Portland Press Limited
206 A.-M. Rosca and A. Burlacu

percentages of Sca-1+ /c-kit+ double-positive cells within studies showed a successful enrichment of haematopoietic
bone marrow have previously been quantified by FACS progenitor cells that were able to repopulate lethally
analysis [11,12]. For example, less than 2% out of 20% irradiated mice. Nijhof and Wierenga isolated cells with
c-kit+ cell populations and 10% Sca-1+ cell populations a diameter between 7.2 and 8.4 m, which sediment
respectively has been found to double-stain for c-kit and at a density of 1.065 g/ml, from spleen; these cells had
Sca-1 in bone marrow mononuclear cells [12]. Another a high capacity to repopulate lethally irradiated mice
study reported that only 0.2% of Lin cells in bone marrow with all haemopoietic precursor cells [21]. In accordance
were Sca-1+ /c-kit+ double-positive [11]. with that study, our flow cytometry results showed a
Because c-kit+ and Sca-1+ cells have low levels preferential distribution of c-kit and Sca-1 progenitor
of granularity, we considered the possibility of using cell markers in a population characterized by a density
this physical property in order to separate these cells. between 1.057 and 1.076 g/ml. By using a discontinuous
Our study showed that a bone marrow subpopulation gradient centrifugation, mouse bone marrow cells were
that is enriched in Sca-1+ and c-kit+ cells can be separated into six fractions (corresponding to 1.050, 1.057,
obtained through density gradient centrifugation and has 1.067, 1.070, 1.076 and 1.083 g/ml), and progenitor cells
the ability to differentiate under specific conditions. This were significantly enriched within fractions IIIV. In any
fraction can be a source for MSCs (CD45 /c-kit /Sca-1+ / case, fraction IV was particularly important because it
PECAM /VEGFR2 /osteocalcin /collagenII cells) that can harboured approx. 2 106 stem/progenitor cells out of a
be used in differentiation and cellular transplantation studies. total of 4.3 106 cells. This population is highly enriched
in stem/progenitor cells compared with the initial bone
marrow population and is valuable in various experiments,
The advantage of using a Percoll gradient to enrich including cellular transplantation studies, which usually need
for cell populations between several thousand and two million cells.
Currently, various techniques based on the physical and
immunochemical characteristics of stem/progenitor cells are
used for their enrichment. Among these, FACS or magnetic Fraction IV as a source of HSCs
bead separation, which are based on cell surface antigen There is always a compromise in choosing the conditions for
expression, are most commonly used for either positive or isolating HSCs between those that provide the highest yield
negative selection [19,20]. These immunochemical methods and those that provide the highest content of progenitor
present the disadvantage of being time-consuming and cells. As a matter of choice, two techniques, namely
expensive because of the use of monoclonal antibodies elutriation and labelling with a selective progenitor marker,
and advanced technology. Although more purity is given are being differentially used for this purpose.
by immunological assays in isolating cells that express a The method described in the present paper provides an
certain surface marker, the resulting cell populations are alternative for the isolation of a haematopoietic-progenitor-
still heterogeneous. That is because no single specific cell-enriched population from bone marrow, in which the
stem cell marker has been identified yet. Likewise, stem physical properties of the progenitor cells are exploited. The
cells are known to express a wide variety of antigens cells in fraction IV, even though they are still heterogeneous,
expressed by many other cell types, and these antigens contain a superior amount of haematopoietic progenitors,
are variably expressed depending on cellular status. On namely 45% of the population, and furthermore, a high
the other hand, mesenchymal stem cell cultures isolated by amount of endothelial progenitor cells. Besides these
immunodepletion of haematopoietic cells have been found haematopoietic progenitor cell populations, this fraction
to exhibit poor growth mainly due to the absence of other still harbours several other cell types, among which are
cells that normally supported their growth by secreting chondroblasts and adipocytes. As a proof of its improvement
paracrine factors. Furthermore, our trials to cultivate a for HSCs, this subpopulation is able to generate an increased
pure population of c-kit+ and Sca-1+ cells isolated by the number of haematopoietic colonies when cultivated under
FACS method also failed due to the absence of survival the appropriate conditions. No prediction can be made
factors provided by supporting cells (results not shown). about the short- or long-term reconstituting properties of
Thus physical separation of bone marrow cells remains a these cells, as they are morphologically indistinguishable
good alternative to immunochemical methods. and such discrimination can be only based on in vivo
Since its introduction in 1977, Percoll has been used functional assays. It is likely that a number of cells have
to isolate several cell types. Its nearly ideal physical reconstituting abilities (as we know that such cells reside
characteristics make it especially useful as a first step within bone marrow); however, we did not perform in vivo
in the enrichment of cell populations before attempting experiments and consequently we have no evidence on this
higher resolution. With the use of this method, several issue.

C 2010 Portland Press Limited
Isolation of a progenitor-enriched population of bone marrow 207

Fraction IV as a source of MSCs


MSCs are usually isolated on the basis of their adherence Funding
to plastic surfaces. Based on this property, MSCs from
different species have been obtained and proven to be This work was supported by grants from the Romanian
able to expand in culture while retaining their multipotent Ministry of Education and Research, IDEA Program [grant
differentiation capacity. Mouse MSC cultures are frequently number 250/2007] and COST (European Co-operation in
contaminated by haematopoietic cells; hence, these cells are Science and Technology) Action BM0602.
more difficult to isolate and expand as compared with MSCs
from other species [13]. Several approaches for isolating
pure populations of mouse MSCs have been reported. For References
instance, immunodepletion of haematopoietic precursors
has been used as a first purification step of bone marrow 1 Pittenger, M. F., Mackay, A. M. and Beck, S. C. (1999) Science
aspirate in order to obtain a more homogenous culture 284, 143147
of MSCs [19,20]. However, the cells have been found to 2 Nervi, B., Link, D. C. and DiPersio, J. F. (2006) J. Cell. Biochem.
exhibit poor growth due to the dramatic down-regulation 99, 690705
of many genes involved in cell proliferation and cell cycle 3 Morrison, S. J. and Spradling, A. C. (2008) Cell 132,
progression as a result of immunodepletion [19]. As an 598611
alternative to immunodepletion, mouse MSC cultures can 4 Friedenstein, A. J., Kailakhyan, R. K., Latsinik, N. V., Panasyuk,
be obtained from diverse strains by incubating total bone A. F. and Keiliss-Borok, I. V. (1974) Transplantation 17,
marrow cells in a high-density culture with medium that 331340
inhibits the growth of haematopoietic cells. In this approach, 5 Jiang, Y., Jahagirdar, B. N., Reinhardt, R. L., Schwartz, R. E.,
the cells are grown under the aforementioned conditions Keene, C. D., Ortiz-Gonzalez, X. R., Reyes, M., Lenvik, T., Lund,
for the first two passages (56 weeks), after which the most T., Blackstad, M. et al. (2002) Nature 418, 4149
easily detached cells are expanded in a low-density culture 6 Tropel, P., Noel, D., Platet, N., Legrand, P., Benabid, A. L. and
[21]. In another reported strategy, cells are plated at 106 Berger, F. (2004) Exp. Cell Res. 295, 395406
cells/cm2 and the medium in the primary culture is changed 7 Baddoo, M., Hill, K., Wilkinson, R., Gaupp, D., Hughes, C.,
frequently concomitant with a diminished trypsinization time Kopen, G. C. and Phinney, D. G. (2003) J. Cell. Biochem. 89,
[22]. In this procedure, a purified population of MSCs can 12351249
be obtained 3 weeks after the initiation of culture (two 8 Hoffman, R., Benz, E., Shattil, S., Furie, B. and Cohen, H. (2008)
passages). In the method described in the present study, a Hematology: basic principles and practice, 5th ed., Churchill
similar pure MSC population (containing CD45 /c-kit /Sca- Livingstone, Philadelphia
1+ /PECAM /VEGFR2 /osteocalcin /collagenII cells) can be 9 Chamberlain, G., Wright, K., Rot, A., Ashton, B. and Middleton,
obtained by cultivating cells from fraction IV for no more J. (2008) PLoS One 3, e2934e2939
than 1 week in DMEM supplemented with 10% MSC- 10 Glassford, A. J., Yue, P., Sheikh, A. Y., Chun, H. J., Zarafshar, S.,
qualified FBS. Chan, D. A., Reaven, G. M., Quertermous, T. and Tsao, P. S.
In conclusion, we designed a method for the (2007) Am. J. Physiol. Endocrinol. Metab. 293, E1590E1596
fractionation of mouse bone marrow through the use 11 Calvi, L. M., Adams, G. B., Weibrecht, K. W., Weber, J. M.,
of a Percoll gradient that resulted in a progenitor- Olson, D. P., Knight, M. C., Martin, R. P., Schipani, E., Divieti, P.,
cell-enriched population. This population of small cells Bringhurst, F. R. et al. (2003) Nature 425, 841846
segregated at a density of 1.070 g/ml and contained both 12 Yamada, Y., Nishimotoa, E., Mitsuyaa, H. and Yonemurab, Y.
mesenchymal and haematopoietic progenitors. These cells (2006) Exp. Hematol. 34, 97106
were able to generate more haematopoietic colonies under 13 Peister, A., Mellad, J. A., Larson, B. L., Hall, B. M., Gibson, L. F.
the appropriate conditions as compared with total bone and Prockop, D. J. (2004) Blood 103, 16621668
marrow. They were also able to produce an MSC line that 14 Anjos-Afonso, F. and Bonnet, D. (2008) Curr. Protoc. Stem
was free of haematopoetic cells in a short time in culture as Cell. Biol. Chapter 2: Unit 2B.3.
compared with classical methods. 15 Zipori, D. (2005) Stem Cells 23, 719726
16 Perez, C., Moreno, S., Summerfield, A., Domenech, N., Alvarez,
B., Correa, C., Alonso, F., Ezquerra, A., Domnguez, J. and
Revilla, C. (2007) J. Immunol. Methods 321, 7079
Acknowledgements 17 Briddell, R. A., Broudy, V. C., Bruno, E., Brandt, J. E., Srour, E. F.
and Hoffman, R. (1992) Blood 79, 31593167
We thank Eugen Andrei, Ioana Manolescu and Dr Emanuel 18 Holmes, C. and Stanford, W. L. (2007) Stem Cells 25,
Dragan for their technical assistance. 13391347


C 2010 Portland Press Limited
208 A.-M. Rosca and A. Burlacu

19 Baddoo, M., Hill, K., Wilkinson, R., Gaupp, D., Hughes, C., 22 Soleimani, M. and Nadri, S. (2009) Nature Protocols 4,
Kopen, G. C. and Phinney, D. G. (2003) J. Cell. Biochem. 89, 102106
12351249
20 Gronthos, S., Graves, S. E., Ohta, S. and Simmons, P. J. (1994)
Blood 84, 41644173 Received 2 December 2009/12 March 2010; accepted 23 March 2010
21 Nijhof, W. and Wierenga, P. K. (1985) Eur. J. Cell. Biol. 39, Published as Immediate Publication 23 March 2010, doi:10.1042/BA20090356

136141


C 2010 Portland Press Limited

Vous aimerez peut-être aussi