ArticlePDF Available

An improved protocol for isolation and culture of mesenchymal stem cells from mouse bone marrow

Authors:

Abstract and Figures

Mesenchymal stem cells (MSCs) from bone marrow are main cell source for tissue repair and engineering, and vehicles of cell-based gene therapy. Unlike other species, mouse bone marrow derived MSCs (BM-MSCs) are difficult to harvest and grow due to the low MSCs yield. We report here a standardised, reliable, and easy-to-perform protocol for isolation and culture of mouse BM-MSCs. There are five main features of this protocol. (1) After flushing bone marrow out of the marrow cavity, we cultured the cells with fat mass without filtering and washing them. Our method is simply keeping the MSCs in their initial niche with minimal disturbance. (2) Our culture medium is not supplemented with any additional growth factor. (3) Our method does not need to separate cells using flow cytometry or immunomagnetic sorting techniques. (4) Our method has been carefully tested in several mouse strains and the results are reproducible. (5) We have optimised this protocol, and list detailed potential problems and trouble-shooting tricks. Using our protocol, the isolated mouse BM-MSCs were strongly positive for CD44 and CD90, negative CD45 and CD31, and exhibited tri-lineage differentiation potentials. Compared with the commonly used protocol, our protocol had higher success rate of establishing the mouse BM-MSCs in culture. Our protocol may be a simple, reliable, and alternative method for culturing MSCs from mouse bone marrow tissues.
Content may be subject to copyright.
ORIGINAL ARTICLE
An improved protocol for isolation and
culture of mesenchymal stem cells from
mouse bone marrow
Shuo Huang
a,b
, Liangliang Xu
a,b
, Yuxin Sun
a,b
, Tianyi Wu
a,b
,
Kuixing Wang
a,b,c
, Gang Li
a,b,c,d,e,
*
a
Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of
Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
b
Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences,
The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
c
Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences,
Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
d
The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System,
The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, Hong Kong, China
e
Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of
Hong Kong, Hong Kong, China
Received 19 May 2014; received in revised form 17 July 2014; accepted 23 July 2014
KEYWORDS
bone marrow;
isolation;
mesenchymal stem
cells;
mouse;
protocol
Summary Mesenchymal stem cells (MSCs) from bone marrow are main cell source for tissue
repair and engineering, and vehicles of cell-based gene therapy. Unlike other species, mouse
bone marrow derived MSCs (BM-MSCs) are difficult to harvest and grow due to the low MSCs
yield. We report here a standardised, reliable, and easy-to-perform protocol for isolation
and culture of mouse BM-MSCs. There are five main features of this protocol. (1) After flushing
bone marrow out of the marrow cavity, we cultured the cells with fat mass without filtering
and washing them. Our method is simply keeping the MSCs in their initial niche with minimal
disturbance. (2) Our culture medium is not supplemented with any additional growth factor.
(3) Our method does not need to separate cells using flow cytometry or immunomagnetic sort-
ing techniques. (4) Our method has been carefully tested in several mouse strains and the re-
sults are reproducible. (5) We have optimised this protocol, and list detailed potential
problems and trouble-shooting tricks. Using our protocol, the isolated mouse BM-MSCs were
strongly positive for CD44 and CD90, negative CD45 and CD31, and exhibited tri-lineage differ-
entiation potentials. Compared with the commonly used protocol, our protocol had higher suc-
cess rate of establishing the mouse BM-MSCs in culture. Our protocol may be a simple, reliable,
and alternative method for culturing MSCs from mouse bone marrow tissues.
Copyright ª2014, Chinese Speaking Orthopaedic Society. Published by Elsevier (Singapore) Pte
Ltd. All rights reserved.
* Corresponding author. Room 904, 9/F, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of
Hong Kong, Shatin, Hong Kong, China.
E-mail address: gangli@cuhk.edu.hk (G. Li).
+MODEL
Please cite this article in press as: Huang S, et al., An improved protocol for isolation and culture of mesenchymal stem cells from mouse
bone marrow, Journal of Orthopaedic Translation (2014), http://dx.doi.org/10.1016/j.jot.2014.07.005
http://dx.doi.org/10.1016/j.jot.2014.07.005
2214-031X/Copyright ª2014, Chinese Speaking Orthopaedic Society. Published by Elsevier (Singapore) Pte Ltd. All rights reserved.
Available online at www.sciencedirect.com
ScienceDirect
journal homepage: http://ees.elsevier.com/jot
Journal of Orthopaedic Translation (2014) xx,1e8
Introduction
Mesenchymal stem cells (MSCs) are multipotent stem cells
that have the potential to self-renew and differentiate into
a variety of specialised cell types such as osteoblasts,
chondrocytes, adipocytes, and neurons [1,2]. MSCs are
easily accessible, expandable, immunosuppressive and they
do not elicit immediate immune responses [3,4]. Therefore,
MSCs are an attractive cell source for tissue engineering
and vehicles of cell therapy.
MSCs can be isolated from various sources such as adi-
pose tissue, tendon, peripheral blood, and cord blood
[5e7]. Bone marrow (BM) is the most common source of
MSCs. MSCs have been successfully isolated and charac-
terised from many species including mouse, rat, rabbit,
dog, sheep, pig, and human [8e12]. Mice are one of the
most commonly used experimental animals in biology and
medicine primarily because they are mammals, small,
inexpensive, easily maintained, can reproduce quickly, and
share a high degree of homology with humans [13]. How-
ever, the isolation and purification of MSCs from mouse
bone marrow is more difficult than other species due to
their heterogeneity and low percentage in the bone marrow
[1,14,15].
Two main stem cell populations and their progenies,
haematopoietic stem cells and BM-MSCs, are the main
residents of bone marrow [1,15]. BM-MSCs are usually iso-
lated and purified through their physical adherence to the
plastic cell culture plate [16]. Several techniques have
been used to purify or enrich MSCs including antibody-
based cell sorting [17], low and high-density culture tech-
niques [18,19], positive and negative selection method
[20], frequent medium changes [21], and enzymatic
digestion approach [22]. However, they all had some short
falls: the standard MSCs culture method based on plastic
adherence has been confirmed to have lower successful
rate [23]; whereas the cell sorting approach reduced the
osteogenic potentials of MSCs [17]. Negative selection
method leads to granulocyteemonocyte lineage cells
reappearing after 1 week of culture [24]. Cells obtained
using a positive selection method show higher proliferation
ability compared with the negative selection method, but
the method was only repeated in the C57B1/6 mice and
failed to repeat in other strains of mice [25]. Frequent
medium change method is inconvenient because it is
required to change the culture medium every 8 hours dur-
ing the first 72 hours of the initial culture [21]. Therefore,
an easy and effective protocol for isolation of mouse BM-
MSCs is needed.
Materials and methods
Reagents
Reagents used included: 0.25% trypsineEDTA (1) with
phenol red; penicillinestreptomycineneomycin (PSN; Life
Technologies, Carlsbad, CA, USA) antibiotic mixture; foetal
bovine serum, qualified, heat-inactivated (Life Technolo-
gies); minimal essential medium (MEM) a, nucleosides,
powder (Life Technologies); and NaHCO
3
(SigmaeAldrich, St
Louis, MO, USA).
Reagent setup
Stock of a-MEM was made up with 1 bag of a-MEM powder
(1 L) and 2.2 g NaHCO
3
in 1000 mL of Milli-Q water, adjusted
to pH 7.2, filtered to sterilise, and stored for 1e2 weeks at
4C. Complete a-MEM medium was a-MEM medium stock
supplemented with 15% foetal bovine serum and 1% PSN,
stored at 4C. Phosphate-buffered saline (PBS) included:
NaCl 8.0 g, KCl 0.2 g, KH
2
PO
4
0.24 g, and Na
2
HPO
4
1.44 g in
1 L Milli-Q water (pH 7.4, sterilised and stored at 4C).
Animals
In this study, two mouse strains (ICR and C57) with different
ages (4 weeks and 8 weeks, males and females) were tested
using our protocol. All mice were purchased from and
housed in a designated and government approved animal
facility at The Chinese University Hong Kong, Hong Kong
SAR, China, in according to The Chinese University Hong
Kong’s animal experimental regulations. All efforts were
made to minimise animal suffering.
BM-MSCS culture protocol
Isolation and culture of mouse BM-MSCs
Mice aged 4 weeks or 8 weeks are terminated by cervical
dislocation and placed in a 100-mm cell culture dish (Bec-
ton Dickinson, Franklin Lakes, NJ, USA), where the whole
body is soaked in 70% (v/v) ethanol for 2 minutes, and then
the mouse is transferred to a new dish (Fig. 1A). Four claws
are dissected at the ankle and carpal joints, and incisions
made around the connection between hindlimbs and trunk,
forelimbs, and trunk. The whole skin is then removed from
the hind limbs and forelimbs by pulling toward the cutting
site of the claw. Muscles, ligaments, and tendons are
carefully disassociated from tibias, femurs, and humeri
using microdissecting scissors and surgical scalpel. Tibias,
femurs, and humeri are dissected by cutting at the joints,
and the bones are transferred onto sterile gauze. Bones are
carefully scrubbed to remove the residual soft tissues
(Fig. 1B), and transferred to a 100-mm sterile culture dish
with 10 mL complete a-MEM medium on ice (Fig. 1C). All
samples are processed within 30 minutes following animal
death to ensure high cell viability. The soft tissues are
completely dissociated from the bones to avoid
contamination.
In a biosafety cabinet, the bones are washed twice with
PBS containing 1% PSN to flush away the blood cells and the
residual soft tissues, then bones are transferred into a new
100-mm sterile culture dish with 10 mL complete a-MEM
medium. The bone is held with forceps and the two ends
excised just below the end of the marrow cavity using
microdissecting scissors (Fig. 1D). A 23-gauge needle
attached to a 5 mL syringe is used to draw 5 mL complete a-
MEM medium from the dish; then the needle is inserted into
the bone cavity. The marrow out is slowly flushed and the
bone cavities washed twice again until the bones become
pale (Fig. 1E). All the bone pieces are removed from the
dish using forceps, leaving the solid mass in the medium,
and the dish is incubated at 37C in a 5% CO
2
incubator for 5
days (Fig. 1F). In order to obtain enough marrow cells, the
2 S. Huang et al.
+MODEL
Please cite this article in press as: Huang S, et al., An improved protocol for isolation and culture of mesenchymal stem cells from mouse
bone marrow, Journal of Orthopaedic Translation (2014), http://dx.doi.org/10.1016/j.jot.2014.07.005
bone cavities are flushed repeatedly until the bones appear
to be pale.
The initial spindle-shaped cells appear on Day 3 in
phase-contrast microscopy, and then culture becomes more
confluent and reaches 70e90% confluence within only 2
days. Cells are washed with PBS twice, and digested with
2.5 mL of 0.25% trypsin for 2 minutes at 37C, then the
trypsin neutralised with 7.5 mL complete a-MEM medium.
The bottom of the plate is flushed using pipet-aid and the
cells transferred to a 15 mL Falcon tube (Becton Dickinson),
which is centrifuged at 800gfor 5 minutes, and the cells
resuspended in a 75 cm
2
cell culture flask (Corning Inc,
Corning, NY, USA) at a split ratio of 1:3. Note: Washing the
cells with PBS prior to digestion is important, as it removes
the residual medium and cell secretion and loosens the
adhesive force of MSCs to the dish. The digestion should be
limited to 2 minutes, as longer digestion is harmful for
MSCs, and could lift non-MSCs from the dish.
Passaging should be performed every 4e6 days at a split
ratio of 1:3. Normally, the cells at Passage 3 contain fewer
macrophages and blood cells, and less fat than those at
Passages 1 and 2, and can be readily used for experiments.
Challenges and possible solutions in mouse BM-MSCs culture
are summarised in Table 1.
The commonly used protocol for establishing (mouse)
BM-MSC culture
The commonly accepted isolation strategy for BM-MSCs was
initially reported by Nadri et al. [16]. Briefly, a mouse is
terminated; tibias, femurs, and humeri are dissected. Both
ends of the bones are removed with sharp scissors. Bone
marrow is flushed out from the bone cavity using plain
culture medium, then filtered through a 70-mm filter mesh,
washed, and resuspended. The dish is then incubated at
37C in a 5% CO
2
incubator. Nonadherent cells are removed
24e72 hours later by changing the medium. When culture
reaches 70e90% confluence, cells are subcultured at a split
ratio of 1:3.
Phenotypic characterisation and cell growth rate of the
mouse BM-MSCs
BM-MSCs at Passage 3 were used for characterisation.
Mesenchymal stem cell markers CD44, CD90, endothelial
cell marker CD31 and haematopoietic marker CD45 were
examined by flow cytometry according to a previously
published paper [26]. The trilineage differentiation abilities
were tested according to previous published protocols
[26,27]. We have used Alizarin red, Oil Red O, and toluidine
blue staining as indicators for osteogenic, adipogenic, and
chondrogenic differentiation according to our previously
published methods [27].
In order to compare the differences between our pro-
tocol and the commonly used protocol for mouse BM-MSCs
culture, we used 16 mice of two different strains (ICR and
C57) with two different ages (4 weeks and 8 weeks, males
and females) and assigned them into four groups as shown
in Table 2. Left/right side of the femurs were chosen for
isolation of BM-MSCs using our protocol and the contralat-
eral femur was then used for isolation of BM-MSCs using the
commonly used method. To calculate the cell growth rate,
when the cultured cells reached about 90% confluence,
they were trypsinised, cell numbers were counted using a
handheld automatic cell counter (Scepter 2.0 Cell Counter,
Merck Millipore, Darmstadt, Germany) and recorded. The
passaged cells were then subcultured into new flasks at a
split ratio of 1:3.
Figure 1 Illustrations of mouse bone marrow cell collection procedures. (A) The mouse was terminated by cervical dislocation,
placed in a 100-mm culture dish, and washed with 70% (vol/vol) ethanol for 2 minutes. (B) Tibias, femurs, and humeri were
dissected; muscles, ligaments, and tendons were removed and the bones transferred onto sterile gauzes. (C) Bones were trans-
ferred to a 100-mm sterile culture dish with 10 mL complete a-minimal essential medium on ice. (D) The dish was transferred into
the biosafety cabinet and washed twice to flush away impurities; the two ends just below the end of the marrow cavity were
excised with microdissecting scissors. (E) A 23-gauge needle was inserted into the bone cavity and used to slowly flush the marrow
out. The bone cavities were washed twice again until the bones became pale. (F) All the bone pieces were removed from the dish
and the fat mass was left in the medium. Then the dish was incubated at 37C in a 5% CO
2
incubator.
Improved isolation and culture of mouse BM-MSCs 3
+MODEL
Please cite this article in press as: Huang S, et al., An improved protocol for isolation and culture of mesenchymal stem cells from mouse
bone marrow, Journal of Orthopaedic Translation (2014), http://dx.doi.org/10.1016/j.jot.2014.07.005
Statistical analysis
All quantitative data were transferred to statistical
spreadsheets and analysed by a commercially available
statistical program SPSS version 16.0 (IBM SPSS Inc., Chi-
cago, IL, USA), one-way analysis of variance were used for
comparison of mean values with p<0.05 considered sta-
tistically significant.
Results
Morphological features of cultured mouse BM-MSCs
Morphological features of the cultured mouse BM-MSCs
using our protocol were similar to those of the BM-MSCs
cultures using the commonly used method. On Day 1, most
of the cells were still mononuclear cells and fat droplets
were frequently seen (Fig. 2A). On Day 2, some spindle-
shaped cells (arrows) appeared among the mononuclear
cells and fat droplets (Fig. 2B). On Day 3, the number of
spindle-shaped cells continued increasing (Fig. 2C). On Day
4, the spindle-shaped cells reached about 60e80% conflu-
ence (Fig. 2D), the cell growth was slower in the flasks using
the commonly used method compared to the new method.
On Day 5, the spindle-shaped cells already formed cell
layers (Fig. 2E) using the new protocol, but this is not seen
in the flasks under the commonly used protocol. Using our
protocol, on Day 5, fibroblast-like cell grew out from a
dense cell nodule (Fig. 2F), and the cells were passaged on
this day; on Day 7, cells reached 100% confluence when left
without passaging, multiple cell layers and dense cell
nodules were formed in some areas of the culture dish
(Fig. 2G and H). By contrast, the cells cultured using the
commonly used method only reach 60e70% confluence at
Day 7. After the cells reached 90% confluence, they were
passed and split; after Passage 3, the cells had uniform
fibroblast-like morphology (Fig. 2I) regardless of the initial
cell culture protocol. Using our protocol, we could obtain
approximately 5 10
7
e1.5 10
8
BM-MSCs from one mouse
bone marrow preparation in about 2 weeks, whereas it
would take at least 3 weeks for the BM-MSCs to reach a
similar cell number using the commonly used protocol.
Confirmation of phenotype and differentiation
capacities of mouse BM-MSCs
Once established in the culture dishes and passaged three
times, the MSCs retain similar phenotypes or differentiation
Table 1 Challenges and possible solutions in mouse BM-MSCs culture.
Problem Possible cause Solution
Few harvested cells
from bone marrow
Incomplete bone marrow
cavity flushing
Repeatedly flush bone cavities until the bones appear to be pale
The bone was broken and
cells leaked out
Carefully dissect bones and dissociate soft tissue from bones
Cells were dead during
harvesting
Prepare the bone marrow within 30 min following animal death,
and keep bones in complete a-MEM medium on ice
Microbial contamination Contaminated during bone
sample harvesting
Wash the mouse body with 70% ethanol for at least 2 min
Avoid bones touching the mouse skin during dissection
Keep bones in complete a-MEM medium with 1% PSN
Contaminated during cell
culture period
Wipe the dish with 70% ethanol prior to transferring it into the
cabinet
Wash bones twice using pipet-aid to flush away impurities,
blood cells and residual soft tissue that slightly connect to
the bone with complete a-MEM medium containing 1% PSN
Cells are not digested
off by trypsin
Cells not washed with PBS
prior to digestion
Wash the cells twice with PBS prior to digestion to remove
any residual serum
Cells grow slowly after
passaging
Trypsin cells for >2 min Digest cells with trypsin for <2 min
The initial total MSC
numbers are low
Do not disturb the cells for the first 3 days and do not passage
the cells until they reach at least 70% confluence
Table 2 Animal group details.
Groups
(Our protocol)
Strain Numbers Age (wk) Femur used
a
Control groups
(Standard protocol)
Femur used
a
I4 ICR 4 4 Random I4-C Contralateral
I8 ICR 4 8 Random I8-C Contralateral
C4 C57 4 4 Random C4-C Contralateral
C8 C57 4 8 Random C8-C Contralateral
a
Left or right side of the femur was randomly used for cell culture using our protocol, and the contralateral femur was then used for
cell culture using the standard protocol.
4 S. Huang et al.
+MODEL
Please cite this article in press as: Huang S, et al., An improved protocol for isolation and culture of mesenchymal stem cells from mouse
bone marrow, Journal of Orthopaedic Translation (2014), http://dx.doi.org/10.1016/j.jot.2014.07.005
potentials regardless of the protocol initially used. Results
showed that the established cells were strongly positive for
MSC markers CD44, CD90 (Fig. 3A and B), and negative for
endothelial cell marker CD31 (Fig. 3C), and the haemato-
poietic cell marker CD45 (Fig. 3D). The isotype control was
negative.
The differentiation capacities of BM-MSCs obtained using
either protocol remain similar. Alizarin red staining
demonstrated that mineralised nodules formed in the BM-
MSCs after 4 weeks under the osteogenic induction
(Fig. 3E). Intracellular Oil-red-O staining showed lipid-rich
vacuoles formation of the mouse BM-MSCs after 2 weeks
adipogenic induction (Fig. 3F). After 3 weeks chondrogenic
induction, the cell pellet was sectioned and stained with
toluidine blue; the positive acidic proteoglycan indicated
the chondrocyte-like cells formation (Fig. 3G).
Comparison of the cell growth rate using the new
protocol versus the commonly used protocol
As shown in Table 3, the successful isolation rate of MSCs
was not strain- or sex- dependent. Cells are able to reach
70e90% confluence from Passage 0e1 using both protocols;
however, cells in Groups I4, I8, C4, and C8 grew significantly
faster than that in control groups, it only takes 5 days when
cells reached 70e90% confluence from P0 to P1 and from P1
to P2 compared with 9 days, and the cell numbers are also
significantly higher than those using the standard culture
protocol (pZ0.037). After the first passage, cells in about
one in four of the cell culture dishes in the control groups
(using the commonly used cell culture method) grew very
slowly or stopped growing, and they were not able to reach
70e90% confluence even after culture for 1 month. These
cells accumulated internal fatty vacuoles, and displayed
the typical large and flat senescent morphology.
Discussion
Isolation of MSCs from bone marrow is far more challenging in
mouse than other species. We compared several reported
isolation strategies and developed a new protocol for stand-
ardised, reliable and easy-to-perform isolation of mouse
MSCs from bone marrow. There are five main features of our
method: First, after flushing bone marrow out of the marrow
cavity, we cultured the cells with fat mass without filtering
them. Our experience showed that the initial phase of culture
is crucial for later yieldof cells. The number of MSCs in mouse
Figure 2 Morphological features of the cultured mouse bone marrow mesenchymal stem cells using our protocol. (A) On Day 1,
most of the cells were still mononuclear and fat droplets (arrows) were frequently seen. (B) On Day 2, some spindle-shaped cells
(arrows) appeared among the mononuclear cells and fat droplets. (C) On Day 3, the numbers of spindle-shaped cells (arrows)
continued increasing. (D) On Day 4, the spindle-shaped cells reached about 60e80% confluence. (E) On Day 5, the spindle-shaped
cells formed cell layers (the circle). (F) On Day 5, fibroblast-like cells (arrows) grew out from a dense cell nodule, and the cells were
passaged on this day. (G, H) On Day 7, cells reached 100% confluence when left without passaging; multiple cell layers and dense
cell nodules were formed in some areas of the culture dish (the circle). (I) At Passage 3, the cells have uniform fibroblast-like
morphology. Scale bar Z200 mm.
Improved isolation and culture of mouse BM-MSCs 5
+MODEL
Please cite this article in press as: Huang S, et al., An improved protocol for isolation and culture of mesenchymal stem cells from mouse
bone marrow, Journal of Orthopaedic Translation (2014), http://dx.doi.org/10.1016/j.jot.2014.07.005
bone marrow is much lower than that of rat or human MSCs
[1], if we filtered the bone marrow, some MSCs attached to
the fat masses will be entrapped onto the filter, thus further
reducing the MSCs yields. Furthermore, BM-MSCs are main-
tained in their niche composing of stromal cells, extracellular
matrix elements, and secreting factors to nurture and regu-
late MSCs self-renewal and differentiation [28]. Our method
is simple, but keeping the MSCs in their initial niche with
minimal disturbanceto allow the initial adjusting time for the
MSCs in culture. Second, the culture medium is not supple-
mented with any additional growth factor. It is reported that
supplement of growth factors in culture may modify MSC
protein synthesis and intracellular trafficking, and affect MSC
proliferation and differentiation potentials [21]. Third, our
method does not need to separate the haematopoietic stem
cells, which are preferentially localised in the endosteal re-
gions of the bone [29] using flow cytometry or immuno-
magnetic sorting techniques. Our method simply use the
culture medium to select the plastic adherent MSCs. Fourth,
this method has been tested in several mouse strains (ICR,
FVB/N, CMV-Luc, C57) as well as in mice with different ages
(2e8 weeks, males and females) and the results are repro-
ducible in all tested mice. The successful isolation rate of
MSCs was not strain- or sex-dependent. Fifth, we have
Figure 3 Confirmation of mesenchymal stem cell (MSC) surface markers and differentiation capacities of mouse bone marrow
(BM)-MSCs. (A, B) Flow cytometry analysis results showed that these cells were positive for MSC markers CD44 (A) and CD90 (B). (C)
Cells were negative for endothelial cell marker CD31. (D) Cells were negative for haematopoietic cell marker CD45. (E) Alizarin red
staining demonstrated that mineralised nodules formed in the BM-MSCs after 4 weeks under the osteogenic induction. (F) Intra-
cellular Oil-red-O staining showed lipid-rich vacuole formation of the mouse BM-MSCs after 2 weeks, adipogenic induction. (G) After
3 weeks’ chondrogenic induction, the cell pellet was sectioned and stained with toluidine blue; the positive acidic proteoglycan
indicated the chondrocyte-like cell formation. Scale bar Z1 mm (F and H) and 100 mm (G).
Table 3 Comparison of cell growth rate using the two protocols among different mice.
Groups Passage 0e1 Passage 1e2
Cells (10
6
) Duration
a
Success rate (%)
b
Cells (10
6
) Duration (d)
a
Success (%)
b
I4-C 2.27 0.06 9 100 3.43 0.08 9 75
I4 2.82 0.11 5 100 4.20 0.15 5 100
I8-C 2.26 0.10 9 100 3.31 0.10 9 75
I8 2.77 0.09 5 100 4.27 0.09 5 100
C4-C 2.27 0.03 9 100 3.32 0.09 9 50
C4 2.85 0.09 5 100 4.21 0.09 5 100
C8-C 2.23 0.10 9 100 3.38 0.06 9 75
C8 2.83 0.16 5 100 4.32 0.09 5 100
a
Duration: defined as the time needed for cells to reach 70e90% confluence after passage.
b
Success rate of cell culture: defined as cells reaching 70e90% confluence after passage.
6 S. Huang et al.
+MODEL
Please cite this article in press as: Huang S, et al., An improved protocol for isolation and culture of mesenchymal stem cells from mouse
bone marrow, Journal of Orthopaedic Translation (2014), http://dx.doi.org/10.1016/j.jot.2014.07.005
optimised our culture protocol, and listed detailed problems
that may arise in cell culture and trouble-shooting tricks
(Table 1). The cells established using our culture method are
mesenchymal stem cells, which are not contaminated with
haematopoietic cell lineages and endothelial cells, and are
able to differentiate into osteoblasts, adipocytes and chon-
drocytes when respectively cultured with osteogenic, adi-
pogenic, and chondrogenic medium.
The current study focuses on the development of a simple
cell culture protocol for mouse BM-MSCs. We used minimal
manipulations during the initial cell harvest (no filtering, no
enzyme digestion). Once the MSCs established in either using
our protocol or the commonly used protocol, the MSCs
remain similar phenotypes or differentiation potentials.
In conclusion, compared with the commonly used mouse
BM-MSCs culture protocol, our protocol results in a higher
success rate of MSCs isolation and establishment in culture;
the cells displayed higher growth rate and maintained the
multipotent differentiation potentials. Our protocol may be
a simple, reliable, and alternative method for culturing
MSCs from mouse bone marrow tissues.
Conflicts of interest
All contributing authors declare no conflicts of interest.
Acknowledgements
This work was supported by a grant from the Hong Kong
Government Research Grant Council, General Research Fund
(CUHK470813), and a grant from the China Shenzhen City
Science and Technology Bureau under the Shenzhen City
Knowledge Innovation Plan, Basic Research Project
(JCYJ20130401171935811) to G.L. This study was also sup-
ported in part by the SMART program, Lui Che Woo Institute of
Innovative Medicine, Faculty of Medicine, The Chinese Uni-
versity of Hong Kong. This research project was made possible
by resources donated by Lui Che Woo Foundation Limited.
References
[1] Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R,
Mosca JD, et al. Multilineage potential of adult human
mesenchymal stem cells. Science 1999;284:143e7.
[2] Sanchez-Ramos JR. Neural cellsderived from adult bone marrow
and umbilical cord blood. J Neurosci Res 2002;69:880e93.
[3] Le Blanc K. Immunomodulatory effects of fetal and adult
mesenchymal stem cells. Cytotherapy 2003;5:485e9.
[4] Kassem M, Abdallah BM. Human bone-marrow-derived
mesenchymal stem cells: biological characteristics and po-
tential role in therapy of degenerative diseases. Cell Tissue
Res 2008;331:157e63.
[5] Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells
in human umbilical cord blood. Br J Haematol 2000;109:
235e42.
[6] Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H,
et al. Human adipose tissue is a source of multipotent stem
cells. Mol Biol Cell 2002;13:4279e95.
[7] Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I,
Fisk NM. Identification of mesenchymal stem/progenitor cells
in human first-trimester fetal blood, liver, and bone marrow.
Blood 2001;98:2396e402.
[8] Meirelles Lda S, Nardi NB. Murine marrow-derived mesen-
chymal stem cell: isolation, in vitro expansion, and charac-
terization. Br J Haematol 2003;123:702e11.
[9] Kadiyala S, Young RG, Thiede MA, Bruder SP. Culture expanded
canine mesenchymal stem cells possess osteochondrogenic
potential in vivo and in vitro. Cell Transplant 1997;6:125e34.
[10] Lennon DP, Caplan AI. Isolation of rat marrow-derived
mesenchymal stem cells. Exp Hematol 2006;34:1606e7.
[11] Ringe J, Kaps C, Schmitt B, Buscher K, Bartel J, Smolian H,
et al. Porcine mesenchymal stem cells. Induction of distinct
mesenchymal cell lineages. Cell Tissue Res 2002;307:321e7.
[12] Caterson EJ, Nesti LJ, Danielson KG, Tuan RS. Human marrow-
derived mesenchymal progenitor cells: isolation, culture
expansion, and analysis of differentiation. Mol Biotechnol
2002;20:245e56.
[13] Crow JFC. C. Little, cancer and inbred mice. Genetics 2002;
161:1357e61.
[14] Dvorakova J, Hruba A, Velebny V, Kubala L. Isolation and
characterization of mesenchymal stem cell population
entrapped in bone marrow collection sets. Cell Biol Int 2008;
32:1116e25.
[15] Zhu H, Guo ZK, Jiang XX, Li H, Wang XY, Yao HY, et al. A
protocol for isolation and culture of mesenchymal stem cells
from mouse compact bone. Nat Protoc 2010;5:550e60.
[16] Nadri S, Soleimani M, Hosseni RH, Massumi M, Atashi A,
Izadpanah R. An efficient method for isolation of murine bone
marrow mesenchymal stem cells. Int J Dev Biol 2007;51:
723e9.
[17] Van Vlasselaer P, Falla N, Snoeck H, Mathieu E. Character-
ization and purification of osteogenic cells from murine
bone marrow by two-color cell sorting using anti-Sca-1
monoclonal antibody and wheat germ agglutinin. Blood
1994;84:753e63.
[18] Eslaminejad MB, Nadri S. Murine mesenchymal stem cell iso-
lated and expanded in low and high density culture system:
surface antigen expression and osteogenic culture minerali-
zation. In Vitro Cell Dev Biol Anim 2009;45:451e9.
[19] Eslaminejad MB, Nikmahzar A, Taghiyar L, Nadri S, Massumi M.
Murine mesenchymal stem cells isolated by low density pri-
mary culture system. Dev Growth Differ 2006;48:361e70.
[20] Baddoo M, Hill K, Wilkinson R, Gaupp D, Hughes C, Kopen GC,
et al. Characterization of mesenchymal stem cells isolated
from murine bone marrow by negative selection. J Cell Bio-
chem 2003;89:1235e49.
[21] Soleimani M, Nadri S. A protocol for isolation and culture of
mesenchymal stem cells from mouse bone marrow. Nat Protoc
2009;4:102e6.
[22] Siclari VA, Zhu J, Akiyama K, Liu F, Zhang X, Chandra A, et al.
Mesenchymal progenitors residing close to the bone surface
are functionally distinct from those in the central bone
marrow. Bone 2013;53:575e86.
[23] Sun S, Guo Z, Xiao X, Liu B, Liu X, Tang PH, et al. Isolation of
mouse marrow mesenchymal progenitors by a novel and reli-
able method. Stem Cells 2003;21:527e35.
[24] Tropel P, Noe
¨l D, Platet N, Legrand P, Benabid AL, Berger F.
Isolation and characterisation of mesenchymal stem cells
from adult mouse bone marrow. Exp Cell Res 2004;295:
395e406.
[25] Nadri S, Soleimani M. Isolation murine mesenchymal stem
cells by positive selection. In Vitro Cell Dev Biol Anim 2007;43:
276e82.
[26] Zhang T, Lee YW, Rui YF, Cheng TY, Jiang XH, Li G. Bone
marrow-derived mesenchymal stem cells promote growth and
angiogenesis of breast and prostate tumors. Stem Cell Res
Ther 2013;4:70.
[27] Chen X, McClurg A, Zhou GQ, McCaigue M, Armstrong MA, Li G.
Chondrogenic differentiation alters the immunosuppressive
property of bone marrow-derived mesenchymal stem cells,
Improved isolation and culture of mouse BM-MSCs 7
+MODEL
Please cite this article in press as: Huang S, et al., An improved protocol for isolation and culture of mesenchymal stem cells from mouse
bone marrow, Journal of Orthopaedic Translation (2014), http://dx.doi.org/10.1016/j.jot.2014.07.005
and the effect is partially due to the upregulated expression
of B7 molecules. Stem Cells 2007;25:364e70.
[28] Kolf CM, Cho E, Tuan RS. Mesenchymal stromal cells. Biology
of adult mesenchymal stem cells: regulation of niche, self-
renewal and differentiation. Arthritis Res Ther 2007;9:204.
[29] Nombela-Arrieta C, Pivarnik G, Winkel B, Canty KJ, Harley B,
Mahoney JE, et al. Quantitative imaging of haematopoietic
stem and progenitor cell localization and hypoxic status in the
bone marrow microenvironment. Nat Cell Biol 2013;15:
533e43.
8 S. Huang et al.
+MODEL
Please cite this article in press as: Huang S, et al., An improved protocol for isolation and culture of mesenchymal stem cells from mouse
bone marrow, Journal of Orthopaedic Translation (2014), http://dx.doi.org/10.1016/j.jot.2014.07.005
... Confluence of cell cultures was about 80% on days 14-21. Then, trypsinization of cells was performed followed by subculturing [18]. ...
... Facial nerve regeneration via BMSCs has proved to be a very promising modality for the treatment of facial nerve disorders [18]. Magnetic targeting technology using nanoparticles as SPIO provided an efficient way of stem cell delivery to the exact injured site, increasing the efficacy of stem cell treatment [19,20]. ...
Article
Full-text available
Aim This study was performed to evaluate neural regenerative capacities of bone marrow stem cells (BMSCs) with or without superparamagnetic iron oxide nanoparticles (SPIONs) as a magnetic targeting tool after neurolysis of the facial nerve (FN) in albino rats. Methods Thirty-eight male albino rats were selected. Two of them were euthanized for normal FN histology assessment. Thirty-six rats were injected with ethanol in the FN nerve for neurolysis induction and assessed one week post-operatively by eye blinking test. Animals were divided into three groups, each containing twelve rats: Group I (positive control) was injected with Dulbecco Modified Eagle's medium (DMEM-F12), group II was injected with BMSCs in DMEM-F12, and group III was injected with BMSCs in DMEM-F12 with poly l-lysine coated SPIONs (0.5 mmol/mL). Monitoring of SPIONs in the rat's body was carried out by MRI. A circular neodymium magnet N52 (0.57 T, 2 × 5 mm) was placed on each rat in group III just below the right ear at the site of surgery to attract SPIONs labeled BMSCs, left in place for 24 h, and then removed. From each group, six rats were euthanized at the end of the 4th and 8th week of treatment, respectively. The right FN trunks were extracted for routine histological examination using H&E stain. Immunohistochemical examination by anti-S100B was performed to characterize the thickness of the myelin sheath formed by the Schwann cells. Ultra-structural examination was performed to study changes in axons, myelin sheaths, and Schwann cells. Results Regeneration of nerve fibers, Schwan cells, and myelin sheaths was better in group II than in groups I and III histologically, immunohistochemically, and ultra-structurally. Conclusion BMSCs alone could ameliorate FN regeneration better than magnetic targeting treatment using BMSCs labeled with SPIONs.
... Rats remaining in groups I, III and IV were anaesthetized and sacrificed on the 7th day of experiment and blood samples were collected for biochemical analysis. Specimens of kidney were obtained and processed for histological and immunohistochemical studies by light and electron microscopes [12,13,14] . ...
... Following euthanasia, the hind limbs were dissected and MSC lines were derived by flushing the tibia and femur with MSC culture media (α-MEM with sodium bicarbonate, without L-glutamine, ribonucleosides and deoxyribonucleosides supplemented with 10% fetal bovine serum, sodium pyruvate, 2 mM L-glutamine, 100 µM L-ascorbic acid, 50 U/mL penicillin and 50 µg/mL streptomycin) and culturing the bone marrow in cell culture dishes at 5% O 2 , 6% CO 2 and a balance of N 2 at 37 • C according to the protocol described in [46]. The cells were passaged twice to reduce the presence of macrophages, blood cells and fat. ...
Article
Full-text available
The impact of age on mesenchymal stromal cell (MSC) characteristics has been well researched. However, increased age is concomitant with increased prevalence of polypharmacy. This adjustable factor may have further implications for the functionality of MSCs and the effectiveness of autologous MSC procedures. We applied hyperspectral microscopy of cell autofluorescence—a non-invasive imaging technique used to characterise cytometabolic heterogeneity—to identify changes in the autofluorescence signals of MSCs from (1) young mice, (2) old mice, (3) young mice randomised to receive polypharmacy (9–10 weeks of oral therapeutic doses of simvastatin, metoprolol, oxycodone, oxybutynin and citalopram), and (4) old mice randomised to receive polypharmacy. Principal Component Analysis and Logistic Regression Analysis were used to assess alterations in spectral and associated metabolic characteristics. Modelling demonstrated that cells from young mice receiving polypharmacy had less NAD(P)H and increased porphyrin relative to cells from old control mice, allowing for effective separation of the two groups (AUC of ROC curve > 0.94). Similarly, cells from old polypharmacy mice were accurately separated from those from young controls due to lower levels of NAD(P)H (p < 0.001) and higher porphyrin (p < 0.001), allowing for an extremely accurate logistic regression (AUC of ROC curve = 0.99). This polypharmacy regimen may have a more profound impact on MSCs than ageing, and can simultaneously reduce optical redox ratio (ORR) and increase porphyrin levels. This has implications for the use of autologous MSCs for older patients with chronic disease.
... The procedures for obtaining MSCs were carried out as previously described. 28,40 In brief, the tibias and femurs were obtained from 10 healthy male mice under sterile conditions. Next, the muscle tissue surrounding the bone was meticulously removed using a scissor and tweezer, ensuring not to damage the bone structure. ...
Article
Full-text available
Background Intracerebral hemorrhage (ICH), a devastating form of stroke, is characterized by elevated morbidity and mortality rates. Neuroinflammation is a common occurrence following ICH. Mesenchymal stem cells (MSCs) have exhibited potential in treating brain diseases due to their anti-inflammatory properties. However, the therapeutic efficacy of MSCs is limited by the intense inflammatory response at the transplantation site in ICH. Hence, enhancing the function of transplanted MSCs holds considerable promise as a therapeutic strategy for ICH. Notably, the iron-quercetin complex (IronQ), a metal-quercetin complex synthesized through coordination chemistry, has garnered significant attention for its biomedical applications. In our previous studies, we have observed that IronQ exerts stimulatory effects on cell growth, notably enhancing the survival and viability of peripheral blood mononuclear cells (PBMCs) and MSCs. This study aimed to evaluate the effects of pretreated MSCs with IronQ on neuroinflammation and elucidate its underlying mechanisms. Methods The ICH mice were induced by injecting the collagenase I solution into the right brain caudate nucleus. After 24 hours, the ICH mice were randomly divided into four subgroups, the model group (Model), quercetin group (Quercetin), MSCs group (MSCs), and pretreated MSCs with IronQ group (MSCs+IronQ). Neurological deficits were re-evaluated on day 3, and brain samples were collected for further analysis. TUNEL staining was performed to assess cell DNA damage, and the protein expression levels of inflammatory factors and the cGAS-STING signaling pathway were investigated and analyzed. Results Pretreated MSCs with IronQ effectively mitigate neurological deficits and reduce neuronal inflammation by modulating the microglial polarization. Moreover, the pretreated MSCs with IronQ suppress the protein expression levels of the cGAS-STING signaling pathway. Conclusion These findings suggest that pretreated MSCs with IronQ demonstrate a synergistic effect in alleviating neuroinflammation, thereby improving neurological function, which is achieved through the inhibition of the cGAS-STING signaling pathway.
... Furthermore, the paracrine role of MSCs was strongly suggested as the next generation of regenerative medicine. This was attributed to the plethora of biologically active molecules constitutively released by MSCs including cytokines, chemokines, growth factors, antioxidants, and miRNA [5,6]. These bioactive molecules predispose a wide range of therapeutic roles like anticancer, antioxidant, antidiabetic, and antiin ammatory [7][8][9][10]. ...
Preprint
Full-text available
The utilization of Mesenchymal stem cells (MSCs)-derived secretome was suggested as a promising alternative in cell-based regenerative therapy. Herein, the MSCs cells were impregnated with a pan-PI3K/Akt/mTOR inhibitor and their secretome was utilized to explore the anticancer and antimetastasis effects against breast cancer. To establish this aim Bone marrow-derived MSCs was treated with 50, 100, or 250 nM Wortmannin (Wort), where the cytotoxic, apoptotic, and autophagic potential of their secretome were investigated in luminal-A breast cancer cells (MCF-7). We found that exposure of MCF-7 to Wort-containing secretome induced both apoptosis and autophagy, whereas prolonged exposure led to massive cell death. Also, Wort-loaded secretome induced nuclear DNA fragmentation and reduced cell metastasis in vitro. These findings were associated with Wort-dependent decrease in the formation of the phosphorylated Akt and mTOR proteins, reduced the expression of their mRNAs, and downregulate of the expression of the catalytic subunit of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K-CA). Taken together, these findings suggested the promising antiproliferative and antimetastasis effects of combining pan-PI3K/Akt/mTOR inhibitors with MSCs-derived secretome in breast cancer.
Article
Full-text available
Augmented osteoclast activity and differentiation can lead to destructive bone diseases, such as arthritis and osteoporosis. Therefore, modulating osteoclastogenesis and differentiation may serve to be a possible strategy for treating such diseases. Tenebrio molitor larvae, also known as mealworms, are considered a good source of protein with nutritional value, digestibility, flavor and functional properties, such as antioxidant, anti-diabetic and anti-obesity effects. However, the role of mealworms in osteoclastogenesis remains poorly understood. The present study therefore investigated the effects of fermented mealworm extract (FME) on receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclastogenesis in bone marrow-derived macrophages (BMMs) whilst also attempting to understand the underlying mechanism, if any. The cells treated with RANKL were used as the negative control. To prepare FME, defatted mealworm powder was fermented with a Saccharomyces cerevisiae strain, and then extracted with fermented alcohol. Cell viability of BMMs isolated from 5-week-old Institute of Cancer Research mice was measured using Cell Counting Kit-8 assay. Subsequently, the effects of FME on osteoclast differentiation were measured using tartrate-resistant acid phosphatase (TRAP) staining. In addition, expression of markers associated with osteoclast differentiation was assessed by reverse transcription-quantitative PCR. Expression of nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) was assessed by western blotting. TRAP staining revealed that FME inhibited osteoclast differentiation in a dose-dependent manner (10-100 µg/ml) without causing cytotoxicity. Specifically, the formation of osteoclasts appear to have been suppressed by FME as indicated by the reduction in the number of TRAP-positive multinucleated cells observed. Furthermore, FME treatment significantly decreased the mRNA expression of c-Fos, whilst also significantly decreasing the expression of NFATc1 on both protein and mRNA levels. c-Fos and NFATc1 are transcription factors that can regulate osteoclast differentiation. FME treatment also reduced the expression of genes associated with osteoclast differentiation and function, including dendritic cell-specific transmembrane protein, osteoclast associated Ig-like receptor, Cathepsin K and TRAP, compared with that in the control group. Subsequently, FME was found to effectively suppress RANKL-induced osteoclast differentiation compared with that by the non-fermented mealworm extract. These findings suggest that FME may confer anti-osteoclastogenic effects, providing insights into its potential application in treatment of osteoporosis.
Article
Full-text available
Bone marrow-derived mesenchymal stem cells (BM-MSCs) play a crucial role in stem cell therapy and are extensively used in regenerative medicine research. However, current methods for harvesting BM-MSCs present challenges, including a low yield of primary cells, long time of in vitro expansion, and diminished differentiation capability after passaging. Meanwhile mesenchymal stem cells (MSCs) recovered from cell banks also face issues like toxic effects of cryopreservation media. In this study, we provide a detailed protocol for the isolation and evaluation of MSCs derived from in vivo osteo-organoids, presenting an alternative to autologous MSCs. We used recombinant human bone morphogenetic protein 2-loaded gelatin sponge scaffolds to construct in vivo osteo-organoids, which were stable sources of MSCs with large quantity, high purity, and strong stemness. Compared with protocols using bone marrow, our protocol can obtain large numbers of high-purity MSCs in a shorter time (6 days vs. 12 days for obtaining passage 1 MSCs) while maintaining higher stemness. Notably, we found that the in vivo osteo-organoid-derived MSCs exhibited stronger anti-replicative senescence capacity during passage and amplification, compared to BM-MSCs. The use of osteo-organoid-derived MSCs addresses the conflict between the limitations of autologous cells and the risks associated with allogeneic sources in stem cell transplantation. Consequently, our protocol emerges as a superior alternative for both stem cell research and tissue engineering.
Article
Full-text available
Regenerative medicine (RM) has emerged as a promising and revolutionary solution to address a range of unmet needs in healthcare, including ophthalmology. Moreover, RM takes advantage of the body’s innate ability to repair and replace pathologically affected tissues. On the other hand, despite its immense promise, RM faces challenges such as ethical concerns, host-related immune responses, and the need for additional scientific validation, among others. The primary aim of this review is to present a high-level overview of current strategies in the domain of RM (cell therapy, exosomes, scaffolds, in vivo reprogramming, organoids, and interspecies chimerism), centering around the field of ophthalmology. A search conducted on clinicaltrials.gov unveiled a total of at least 209 interventional trials related to RM within the ophthalmological field. Among these trials, there were numerous early-phase studies, including phase I, I/II, II, II/III, and III trials. Many of these studies demonstrate potential in addressing previously challenging and degenerative eye conditions, spanning from posterior segment pathologies like Age-related Macular Degeneration and Retinitis Pigmentosa to anterior structure diseases such as Dry Eye Disease and Limbal Stem Cell Deficiency. Notably, these therapeutic approaches offer tailored solutions specific to the underlying causes of each pathology, thus allowing for the hopeful possibility of bringing forth a treatment for ocular diseases that previously seemed incurable and significantly enhancing patients’ quality of life. As advancements in research and technology continue to unfold, future objectives should focus on ensuring the safety and prolonged viability of transplanted cells, devising efficient delivery techniques, etc.
Article
Full-text available
Introduction Mesenchymal stem cells (MSCs) are known to migrate to tumor tissues. This behavior of MSCs has been exploited as a tumor-targeting strategy for cell-based cancer therapy. However, the effects of MSCs on tumor growth are controversial. This study was designed to determine the effect of MSCs on the growth of breast and prostate tumors. Methods Bone marrow-derived MSCs (BM-MSCs) were isolated and characterized. Effects of BM-MSCs on tumor cell proliferation were analyzed in a co-culture system with mouse breast cancer cell 4T1 or human prostate cancer cell DU145. Tumor cells were injected into nude mice subcutaneously either alone or coupled with BM-MSCs. The expression of cell proliferation and angiogenesis related proteins in tumor tissues were immunofluorescence analyzed. The angiogenic effect of BM-MSCs was detected using tube formation assay. The effects of the crosstalk between tumor cells and BM-MSCs on expression of angiogenesis related markers were examined by immunofluorescence and real-time PCR. Results Both co-culturing with mice BM-MSCs (mBM-MSCs) and treatment with mBM-MSCs-conditioned medium enhanced the growth of 4T1 cells. Co-injection of 4T1 cells and mBM-MSCs into nude mice led to increased tumor size compared with injection of 4T1 cells alone. Similar experiments using DU145 cells and human BM-MSCs (hBM-MSCs) instead of 4T1 cells and mBM-MSCs got consistent results. Compared with tumors induced by injection of tumor cells alone, blood vessel area was greater in tumors from co-injection of tumor cells with BM-MSCs, which correlated with decreased central tumor necrosis and increased tumor cell proliferation. Furthermore, both conditioned medium from hBM-MSCs alone and co-cultures of hBM-MSCs with DU145 cells were able to promote tube formation ability of human umbilical vein endothelial cells (HUVEC). When hBM-MSCs exposed to DU145 cells environment, the expression of markers associated with neovascularization (macrophage inflammatory protein-2 (MIP-2), vascular endothelial growth factor (VEGF), transforming growth factor-beta (TGF-beta) and interleukin-6 (IL6) were increased. Conclusions These results indicate that BM-MSCs promote tumor growth and suggest that the crosstalk between tumor cells and BM-MSCs increased the expression of pro-angiogenic factors, which may have induced tumor cell proliferation and angiogenesis thereby increasing solid tumor growth.
Article
Full-text available
The existence of a haematopoietic stem cell niche as a spatially confined regulatory entity relies on the notion that haematopoietic stem and progenitor cells (HSPCs) are strategically positioned in unique bone marrow microenvironments with defined anatomical and functional features. Here, we employ a powerful imaging cytometry platform to perform a comprehensive quantitative analysis of HSPC distribution in bone marrow cavities of femoral bones. We find that HSPCs preferentially localize in endosteal zones, where most closely interact with sinusoidal and non-sinusoidal bone marrow microvessels, which form a distinctive circulatory system. In situ tissue analysis reveals that HSPCs exhibit a hypoxic profile, defined by strong retention of pimonidazole and expression of HIF-1α, regardless of localization throughout the bone marrow, adjacency to vascular structures or cell-cycle status. These studies argue that the characteristic hypoxic state of HSPCs is not solely the result of a minimally oxygenated niche but may be partially regulated by cell-specific mechanisms.
Article
Full-text available
Unlike humans, mouse bone marrow-derived mesenchymal stem cells (MSCs) cannot be easily harvested by adherence to plastic owing to the contamination of cultures by hematopoietic cells. The design of the protocol described here is based on the phenomenon that compact bones abound in MSCs and hematopoietic cells exist in the marrow cavities and the inner interfaces of the bones. The procedure includes flushing bone marrow out of the long bones, digesting the bone chips with collagenase type II, deprivation of the released cells and culturing the digested bone fragments, out of which fibroblast-like cells migrate and grow in the defined medium. The entire technique requires 5 d before the adherent cells are readily passaged. Further identification assays confirm that these cells are MSCs. We provide an easy and reproducible method to harvest mouse MSCs that does not require depletion of hematopoietic cells by sorting or immunomagnetic techniques.
Article
Osteogenic cells were sorted from bone marrow of 5-fluorouracil (5-FU)- treated mice based on light scatter characteristics, Sca-1 expression, and their binding to wheat germ agglutinin (WGA). Four sort gates were established using forward (FSC) and perpendicular (SSC) light scatter and were denominated as FSChigh SSClow, FSClow SSChigh, FSClow SSClow, and FSChigh SSChigh cell. Cells from the FSChigh SSChigh gate, but not from the other gates, synthesized alkaline phosphatase, collagen, and osteocalcin and formed a mineralized matrix in culture. The number of osteoprogenitor cells was significantly enriched after depleting the 5- FU bone marrow from cells of the lymphoid and myeloid lineage, eg, T cells, B cells, natural killer cells, granulocytes, macrophages, and erythrocytes. Approximately 95% of the FSChigh SSChigh cell population of this “lineage-negative” (Lin-) marrow expressed the Sca-1 antigen (Sca-1+) and bound WGA. Three additional sort windows were established based on WGA binding intensity and were denominated as Sca-1+ WGAdull, Sca-1+ WGAmedium, and Sca-1+ WGAbright. Cells from the Sca-1+ WGAbright gate, but not from the other gates, synthesized bone proteins and formed a mineralized matrix. However, they lost this capacity upon subcultivation. Further immunophenotypic characterization showed that FSChigh SSChigh Lin- Sca-1+ WGAbright cells expressed stromal (KM16) and endothelial (Sab-1 and Sab-2) markers, but not hematopoietic surface markers such as c-kit and Thy1.2. Sorted FSChigh SSChigh Lin- Sca-1+ WGAbright cells form three-dimensional nodules that stain with the von Kossa technique and contain osteoblast and osteocyte-like cells.
Article
Human mesenchymal stem/progenitor cells (MSCs) have been identified in adult bone marrow, but little is known about their presence during fetal life. MSCs were isolated and characterized in first-trimester fetal blood, liver, and bone marrow. When 106 fetal blood nucleated cells (median gestational age, 10+2 weeks [10 weeks, 2 days]) were cultured in 10% fetal bovine serum, the mean number (± SEM) of adherent fibroblastlike colonies was 8.2 ± 0.6/106 nucleated cells (69.6 ± 10/μL fetal blood). Frequency declined with advancing gestation. Fetal blood MSCs could be expanded for at least 20 passages with a mean cumulative population doubling of 50.3 ± 4.5. In their undifferentiated state, fetal blood MSCs were CD29+, CD44+, SH2+, SH3+, and SH4+; produced prolyl-4-hydroxylase, α-smooth muscle actin, fibronectin, laminin, and vimentin; and were CD45−, CD34−, CD14−, CD68−, vWF−, and HLA-DR−. Fetal blood MSCs cultured in adipogenic, osteogenic, or chondrogenic media differentiated, respectively, into adipocytes, osteocytes, and chondrocytes. Fetal blood MSCs supported the proliferation and differentiation of cord blood CD34+cells in long-term culture. MSCs were also detected in first-trimester fetal liver (11.3 ± 2.0/106 nucleated cells) and bone marrow (12.6 ± 3.6/106 nucleated cells). Their morphology, growth kinetics, and immunophenotype were comparable to those of fetal blood-derived MSCs and similarly differentiated along adipogenic, osteogenic, and chondrogenic lineages, even after sorting and expansion of a single mesenchymal cell. MSCs similar to those derived from adult bone marrow, fetal liver, and fetal bone marrow circulate in first-trimester human blood and may provide novel targets for in utero cellular and gene therapy.
Article
Long bone is an anatomically complicated tissue with trabecular-rich metaphyses at two ends and cortical-rich diaphysis at the center. The traditional flushing method only isolates mesenchymal progenitor cells from the central region of long bones and these cells are distant from the bone surface. We propose that mesenchymal progenitors residing in endosteal bone marrow that is close to the sites of bone formation, such as trabecular bone and endosteum, behave differently from those in the central bone marrow. In this report, we separately isolated endosteal bone marrow using a unique enzymatic digestion approach and demonstrated that it contained a much higher frequency of mesenchymal progenitors than the central bone marrow. Endosteal mesenchymal progenitors express traditional mesenchymal stem cell markers and are capable of multi-lineage differentiation. However, we found that mesenchymal progenitors isolated from different anatomical regions of the marrow did exhibit important functional differences. Compared to their central marrow counterparts, endosteal mesenchymal progenitors have superior proliferative ability with reduced expression of cell cycle inhibitors. They showed greater immunosuppressive activity in culture and in a mouse model of inflammatory bowel disease. Aging is a major contributing factor for trabecular bone loss. We found that old mice have a dramatically decreased number of endosteal mesenchymal progenitors compared to young mice. Parathyroid hormone (PTH) treatment potently stimulates bone formation. A single PTH injection greatly increased the number of endosteal mesenchymal progenitors, particularly those located at the metaphyseal bone, but had no effect on their central counterparts. In summary, endosteal mesenchymal progenitors are more metabolically active and relevant to physiological bone formation than central mesenchymal progenitors. Hence, they represent a biologically important target for future mesenchymal stem cell studies.
Article
Marrow culture from mice has been reported to be overgrown by non-mesenchymal cells. In almost all protocols for isolation of murine mesenchymal stem cells (MSCs), high density culture systems have been employed. Since MSCs are colonogenic cells, the initiating cell seeding density may have significant impact on their cultures. This subject was explored in this study. For this purpose, the bone marrow cells from NMRI mice were plated at 2.5 x 10(6) cells/cm(2) and upon confluency were reseeded as either low density (50 cells/cm(2)) or high density (8 x 10(4) cells/cm(2)) cultures. The cells were expanded through an additional subculture and the passage 2 cells as a product of two culture systems were statistically compared with respect to their surface antigen profiles and osteogenic culture mineralization. While low density culture grew with multiple colony formation, there were no distinct colonies in high density cultures. In contrast to high density cultures, passage 2 cells from low density system possessed typical homogenous fibroblastic morphology. Some cells from high density system but not the low density cultures expressed hematopoietic and endothelial cell markers including CD135, CD34, CD31, and Vcam surface antigens. Furthermore, osteogenic cultures from low density system displayed significantly more mineralization than those from high density system. Taken together, it seems that low density culture system resulted in more purified MSC culture than its counterpart as high density culture system.