Sie sind auf Seite 1von 8

STEM CELLS AND DEVELOPMENT

Volume 18, Number 10, 2009


© Mary Ann Liebert, Inc.
DOI: 10.1089/scd.2008.0223

Isolation and Differentiation of Chicken Mesenchymal Stem


Cells From Bone Marrow

Mahesh Khatri,1,* Timothy D. O’Brien,2 and Jagdev M. Sharma1,†

Mesenchymal stem cells (MSCs) are multipotent progenitor cells found in bone marrow that have the capac-
ity of differentiating into bone, cartilage, fat, muscle, and other tissues. Chicken MSCs were isolated from 1- to
14-day-old chickens. Microscopically, the cultured cells showed morphology resembling fibroblasts and divided
actively. Chicken MSCs expressed the transcription factors PouV, Sox2, and Nanog, which have been shown to be
critical for stem cell self-renewal and pluripotency. The multilineage differentiation potential of chicken MSCs
was revealed by their ability to undergo adipogenic, osteogenic, and chondrogenic differentiation. Like mam-
malian MSCs, chicken MSCs also had immunoregulatory activity and inhibited in vitro mitogenic response of
T cells. The inhibition of mitogenic response of T cells correlated with the production of nitric oxide (NO) in
cultures containing MSCs and T cells. Our data show for the first time that MSCs can be isolated from postnatal
chicken bone marrow and these cells are capable of in vitro multiplication and multilineage differentiation, thus
making them a suitable model in the field of stem cell research.

Introduction
teristics indicate that MSCs can be used as powerful tools in

M esenchymal stem cells (MSCs) were first isolated from


bone marrow as precursors for fibroblasts or stromal
cells. MSCs, also known as plastic-adherent marrow cells or
reconstructive medicine. Pluripotent stem cells are an attrac-
tive tool for tissue engineering and regenerative medicine.
MSCs isolated from humans as well as from other mam-
bone marrow stromal cells, have potential to differentiate malian species have immunoregulatory function and inhibit
into cells with mesenchymal, mesodermal, neuroectodermal, inflammation and immunological responses both in vitro
or endodermal lineage [1]. MSCs have been isolated from sev- and in vivo. MSCs suppress T-cell proliferation probably via
eral species, including human [2], mouse [3], rat [4], dog [5], mechanisms that are independent of major histocompati-
baboon, pig, sheep, goat, rabbit [6], and cat [7]. MSCs have bility complex (MHC). Several soluble factors such as trans-
been isolated from embryonic chicks [8,9]; however, this is forming growth factor-β (TGF-β), hepatocyte growth factor,
the first report of isolation of MSCs from postnatal chickens. indoleamine 2,3-dioxygenase (IDO), nitric oxide (NO), and
MSCs constitute a small population (~0.001–0.01% nucle- prostaglandin E2 (PGE2) have been reported to mediate
ated cells) of adult human bone marrow cells. MSCs have T-cell suppression by MSCs [10]. MSCs improve the outcome
been isolated from several tissues including adipose tissue, of allogeneic transplantation by promoting hematopoietic
skeletal muscle, synovium, spleen, thymus, blood, lung, engraftment [17] and also reduce graft-versus-host disease
fetal blood, and amniotic fluid [10]. These cells have the [18,19]. In vivo infusion of MSCs improved the course of
ability to produce cytokines and growth factors that support chronic autoimmune encephalomyelitis in multiple sclerosis
and regulate haematopoiesis [11,12]. Several studies using a and other neurodegenerative disorders [20].
variety of animal models have shown that MSCs may have a Chicken is the first farm animal with a completely se-
role in the repair and regeneration of infarcted myocardium, quenced genome. Because of its in ovo embryonic develop-
damaged bone, tendon, and cartilage [13–16]. These charac- ment rather than in utero, chicken is an important model for

1
Department of Veterinary and Biomedical Sciences and 2Department of Veterinary Population Medicine, College of Veterinary
Medicine, University of Minnesota, Saint Paul, Minnesota.
*Current affiliation: Department of Food Animal Health Research Program, Ohio Agricultural Research and Development Center,
Ohio State University, Wooster, Ohio.

Current affiliation: The Biodesign Institute, Arizona State University, Tempe, Arizona.

1485
1486 KHATRI, O’BRIEN, AND SHARMA

embryology and development studies. Chicken serves as a media was completely replaced every 3–4 days. Adipogenic
model organism for the study of viruses and cancer. Rous potential was assessed by Oil Red O staining. The cells were
sarcoma virus, the first tumor virus, and oncogene (src) were washed with PBS and fixed with 10% formalin for 30 min at
identified in the chicken. The chicken immune system pro- room temperature. Then the cells were incubated with fil-
vided the first indication of the distinctions between T and B tered 0.3% Oil Red O (Sigma-Aldrich, St. Louis, MO) in 60%
cells, with the B-cell nomenclature based on the avian bursa isopropanol for 30 min. After three washes in PBS, the cells
of Fabricius [21]. Thus, chicken is an important model system were photographed.
for studies of evolution, embryology, immunology, oncology, Osteogenic differentiation. Osteogenic differentiation was
cell biology, virology, and gene regulation. assayed by the method of Pittenger et al. [2]. In brief, MSCs
In the present study, we have, for the first time, isolated were seeded in 24-well plates at a density of ~104 cells/well
and characterized MSCs from hatched specific pathogen-free and cultured in expansion media until the cells reached 90%–
chickens. Chicken MSCs may be useful as a model system 100% confluence. Osteogenic differentiation was induced by
for studies on development, physiology, and disease. culturing the cells for 21 days in medium consisting of 100 nM
dexamethasone, 10 mM β-glycerophosphate (Sigma-Aldrich,
Materials and Methods St. Louis, MO), 0.05 mM l-ascorbic acid-2-phosphate (Sigma-
Aldrich), and 10% FBS in DMEM. Fresh media was fed to cul-
Isolation and expansion of MSCs tures every 3–4 days. Osteogenesis as indicated by calcium
deposition was evaluated by von Kossa staining. The cells
Femur bones were obtained from 1- to 14-day-old spe-
were washed with PBS, fixed with 10% formalin for 30 min at
cific pathogen-free chickens. MSCs were isolated by previ-
room temperature, washed three times with distilled water,
ously described methods [2,22]. In brief, the tip of each bone
and incubated in 5% silver nitrate (Sigma-Aldrich) for 45 min.
was removed and the marrow was harvested by inserting
The cells were washed three times with distilled water and
a syringe needle into one end of the bone and flushing
incubated with sodium thiophosphate (5%) (Sigma-Aldrich)
with Dulbecco’s modified Eagle’s medium (DMEM; Gibco,
for 2 min. Finally, the cells were washed three times with dis-
Carlsbad, CA). The bone marrow cells were filtered through
tilled water and photographed.
a 70-μm nylon mesh filter (BD, Falcon, San Jose, CA) and
Chondrogenic differentiation. Chondrogenic differen-
mononuclear cells were obtained by density gradient centri-
tiation was assayed by the method of Pittenger et al. [2]
fugation over Ficoll-Hypaque (gradient density 1.090). Cells
with minor modifications. The chondrogenic induction
(1–5 × 105/cm2) were plated in 25-cm2 cell culture flasks
of chicken MSCs was examined using high-density pellet
in DMEM containing 10% fetal bovine serum (FBS; Atlas
cultures (2 × 105 cells/pellet, 500 × g for 5 min) in DMEM
Biological, CO), 2 mm l-glutamine (Gibco, USA), and 1% an-
medium supplemented with 50 mM ascorbic acid, 0.5 mg/
tibiotic solution (Sigma, St. Louis, MO). Cultures were incu-
mL insulin (Sigma). Recombinant human transforming
bated at 37°C in a humidified atmosphere containing 95%
growth factor-β1 (Invitrogen, Carlsbad, CA) was added to
air and 5% CO2. The nonadherent cells were removed after
the pellet cultures at a final concentration of 10 ng/mL. The
72 h of culture. When primary cultures became nearly con-
medium was replaced every 2–3 days for 21 days. Sulfated
fluent, the cells were detached with 0.025% trypsin contain-
glycosaminoglycans were detected by 1% Alcian blue stain-
ing 0.02% EDTA and used for future experiments.
ing. The pellet was washed with PBS, fixed with 10% for-
malin for 30 min at room temperature, washed three times
Colony-forming unit-fi broblast (CFU-F) assay with distilled water, and incubated in 1% Alcian blue (pH 1)
The colony-forming potential of MSCs was tested by plat- (Sigma-Aldrich) overnight. The cell pellet was washed three
ing 100 or 1,000 cells in a 60-mm2 cell culture dish. After 7–8 times with distilled water and photographed.
days of incubation at 37°C, the cells were fixed and stained
with 1% crystal violet in methanol for 30 min. All visible col- RNA extraction and RT-PCR
onies greater than 3 mm were counted [22].
RT-PCR was performed to examine the expression of
transcription factors and to confirm gene expression indic-
Proliferation of MSCs
ative of cell differentiation [2,23]. RNA was isolated using
The proliferation rate of chicken MSCs was determined the TRIzol total RNA isolation reagent (Life Technologies,
by cell count. For cell count: a total of 1 × 104 cells were plated Rockville, MD). The extracted RNA (1 μg) was reverse tran-
in a 24-well plate and a total of three replicas were prepared scribed by adding 5 mM of random hexamer oligonucle-
for each observation. At intervals, the cells were trypsinized otides (Life Technologies), 200 units of SuperScript reverse
and a total cell count was determined by Trypan blue dye transcriptase (Life Technologies), 0.5 mM deoxyribonu-
exclusion method. cleotide triphosphates (dNTP) (Life Technologies), and 10
mM dithiothreitol (Life Technologies). The cDNA was then
Differentiation assays amplified by PCR using primer sequences shown in Table 1.
After PCR, 10 μL of the reaction mixture was subjected to
Adipogenic differentiation. Adipogenic differentiation was electrophoresis on a 1.5% agarose gel, and the PCR products
assayed by the method of Pittenger et al. [2]. In brief, ~104 cells/ were visualized by ethidium bromide staining.
well in 24-well plates were cultured in expansion media until
the cells reached 70%–80% confluence. Adipogenic differen- T-cell proliferation assay
tiation was induced by culturing confluent MSCs cultures in
DMEM supplemented with 10% FBS, 1 μM dexamethasone, Splenocytes were prepared by centrifugation over
10 μg/mL insulin, 0.2 mM indomethacin for 21 days. The Ficoll-Hypaque gradients [24]. The cells were suspended in
CHICKEN MESENCHYMAL STEM CELLS FROM BONE MARROW 1487

Table 1. PCR Primer Sequences

PCR Accession
Gene Primers sequence (5′–3′) product (bp) no.

β-Actin Forward: CATCACCATTGGCAATGAGAGG 353 L08165


Reverse: GCAAGCAGGAGTACGATGAATC
CD44 Forward: GGTTTTATAGTGGGGCATATTGTTATCCC 700 AF153205
Reverse: TTAACCGCGATGCACACGGC
CD90 Forward: GGTCTACATGTGCGAGCTGA 471 NM 204381
Reverse: AAAGCTAAGGGGTGGGAGAA
CD45 Forward: CACTGGGAATCGAGAGGAAA 574 NM 204417
Reverse: CTGGTCTGGATGGCACTTTT
CD105 Forward: ACGGATGACACCATGGAAAT 704 AY702002
Reverse: ATGAGGAAGGCTCCAAAGGT
PouV Forward: AAATGTGTGAAGCCCAGTCC 401 DQ867024
Reverse: TTGTGGAAAGGTGGCATGTA
Sox2 Forward: TCCGGCGGTAATAATAGCAG 312 U12532
Reverse: TTGCTGATCTCCGAGTTGTG
Nanog Forward: CAGCAGACCTCTCCTTGACC 371 DQ867025
Reverse: CCAGATACGCAGCTTGATGA
PPAR-γ Forward: TACATAAAGTCCTTCCCGCTGACC 401 AF163811
Reverse: TCCAGTGCGTTGAACTTCACAGC
aP2 Forward: GAGTTTGATGAGACCACAGCAGA 312 AF432507
Reverse: ATAACAGTCTCTTTGCCATCCCA
Osteopontin Forward: TGCCAGGAAGCTCATTGAGGATG 371 M59182
Reverse: GCGTCTACATTTACAAACACACGTC
Collagen II Forward: GCAGAGACCATCAACGGCGGT 333 AY046949.1
Reverse: CAGGCGCGAGGTCTTCTGCGA

triplicate in 100 µL RPMI 1,640 medium supplemented with Statistical analysis


5% FBS (Sigma), 2 mM l-glutamine (Invitrogen, Carlsbad,
CA), 0.1 mg/mL streptomycin, and 100 U/mL penicillin G Statistical analysis was performed by analysis of vari-
(Invitrogen) in 96-well plate in the presence or absence of ance (ANOVA) followed by comparison of individual means
MSCs. The cultures were stimulated or unstimulated with by Tukey’s test. P < 0.05 was considered to be statistically
mitogen (ConA, Phorbol 12-myristate 13-acetate (PMA), and significant.
ionomycin) and incubated for 48 h at 37°C. The cells were
pulsed with [3H] thymidine during the last 5 h of incubation. Results
Lymphoproliferation was measured as counts per minute by
a Matrix 9600 beta counter (Packard Instrument Co., Meriden, Cell culture
CT). All experiments were performed in triplicates.
ConA was purchased from Sigma (St. Louis, MO). Con A We isolated chicken MSCs from the bone marrow based
was used at 5 μg/mL. PMA and ionomycin were from Sigma on their characteristic property of attachment to a plastic
and were used at concentrations of 50 ng/mL and 1 μg/mL, surface. MSCs showed a fibroblast-like morphology. The ini-
respectively. tial adherent spindle-shaped cells appeared as individual
cells on the third day of culture. Within 2 weeks, the cells
reached 70%–80% confluency (Fig. 1A). To confirm that the
Measurement of NO production isolated cells had MSCs characteristics, we examined their
NO production was determined by measurement of differentiation capacity and immune-modulating capabili-
nitrite in the medium as described earlier [25]. In brief, 50 ties as shown below.
μL of the spent medium from the cultures mentioned in the
T-cell proliferation assay was mixed with an equal volume
CFU assay
of 1:1 mixture of 1% sulfanilamide in water and 0.1% N-1-
naphthylethylenediamine dihydrochloride in 5% phospho- MSCs isolated from humans and other animal species
ric acid. The absorbance was then read at 570 nm. Sodium have the ability to self-renew and propagate. The CFU assay
nitrite dissolved in the culture medium was used to con- was used to assess the proliferation and colony-forming
struct the standard curve. NO concentration was measured potential of chicken MSCs. Chicken MSCs formed 30 ± 8
in micromolars (μM). All experiments were performed in and 65 ± 10 colonies when plated at 100 and 1,000 cells per
triplicates. 60-mm culture dish, respectively (Fig. 1B).
1488 KHATRI, O’BRIEN, AND SHARMA

A B SF P2 P8
PouV

Sox2

Nanog

β-Actin

FIG. 2. Transcription factor expression in chicken mesen-


C D chymal stem cells (MSCs). Total cellular RNA was extracted
MSCs Cont. from MSCs and subjected to RT-PCR by using specific prim-
30 CD44 ers to PouV, Sox2, Nanog, or β-actin. PouV, Sox2, and Nanog
* gene expression was detected up to passage 8. Total RNA
Cells (10×e4)

25
* CD45
20 prepared from chicken skin fibroblasts (SF) was used as a
15 CD90 negative control.
10
5 CD105
0
1 2 3 4 5 6 β-Actin
been shown to regulate the pluripotency and self-renewal
Days of chicken embryonic stem cells [30]. As shown in Figure 2,
like mammalian MSCs [23,31], PouV, Nanog, and Sox2 gene
FIG. 1. Morphology, colony formation, and proliferation transcripts were expressed in chicken MSCs for as long as
potential of chicken mesenchymal stem cells (MSCs). (A) eight passages in vitro (highest passage level tested).
Morphology of in vitro-expanded chicken MSCs obtained
from bone marrow. Culture-expanded chicken MSCs dem-
Differentiation of chicken MSCs
onstrate the spindle-shaped fibroblastic morphology. (B)
Colony-forming assay. A single cell can be expanded to To examine the multipotentiality of chicken MSCs, the
generate clones and form colonies (CFU) that are shown by cells were cultured in the media specific for the induction of
crystal violet staining. (C) Proliferation potential of MSCs. adipocytes and osteocytes.
MSCs were seeded in a 24-well plate and their proliferation For the generation of adipocytes, 70%–80% confluent
was measured over 6-day period by counting the viable cells cultures were incubated with adipocytes induction media.
by Trypan blue dye. *Significantly different from Day 1; P < After 21 days of incubation, differentiation of MSCs into
0.05. (D) Phenotype of chicken MSCs. Reverse transcription- adipocytes was determined by staining with Oil Red O.
polymerase chain reaction was performed using total RNA Differentiated cells contained multiple lipid vacuoles (Fig. 3A
extracted from passage 8 cells with specific primers, which and B). The accumulation of lipid vacuoles in the cells was
are shown in Table 1. The left column shows the results with first detected between 48 and 72 h after the addition of adipo-
total RNA from MSCs. Total RNA prepared from normal genic medium. The expression of adipogenic-specific genes,
chicken spleen cells was used as a control (right column). aP2 and PPAR-γ, was induced in the adipocytes, thereby
confirming the adipogenic differentiation of MSCs (Fig. 3E).
Following incubation of MSCs in osteogenic media,
The proliferation potential of MSCs is shown in Figure tightly packed colonies forming nodule-like structures were
1C. These cells are continuously in culture for >8 months. observed. Deposition of calcium in these cells was dem-
These data indicate that chicken MSCs are capable of self- onstrated by staining with von Kossa stain (Fig. 3C). The
renewal and have high proliferation property. osteocyte-specific gene, osteopontin was also up-regulated
To examine the phenotype of chicken MSCs, we per- in differentiated cells (Fig. 3F).
formed RT-PCR using specific primers for various mark- After 14 days of culture, MSCs exposed to TGF-β1 formed
ers (Table 1; Fig. 1D). The cells showed positive reactions a compact and homogeneous pellet structure, showing an
for CD44, CD90 (Thy-1), and CD105 (Endoglin), but did not increased cellular density. Alcian blue staining revealed a
express CD45 indicating that these cells originated from homogeneous deposition of proteoglycan within the whole
mesenchymal lineage cells. section of the pellet culture (Fig. 3D). The differentiated cells
showed gene expression of collagen II (Fig. 3G).
Expression of PouV, Sox2, and Nanog transcription
factors in chicken MSCs Characteristics of T-cell suppression by MSCs
Several transcription factors are known to regulate the MSCs isolated from humans, mice, and other animal spe-
development and differentiation of MSCs. In mammals, cies suppress T-cell proliferation. Likewise, chicken MSCs
MSCs pluripotency is under the control of transcription fac- also inhibited ConA-induced T-cell proliferation (Fig. 4A).
tors that include Oct4 [26], Nanog [27], Sox2 [28], and FoxD3 We next induced T-cell proliferation using a combination
[29]. Oct4, the octomer-binding transcription factor 4, is an of PMA and ionomycin. PMA and ionomycin induce the dif-
important binding transcription factor present in undiffer- ferentiation and expansion of immune cells by activating
entiated embryonic and adult stem cells with a high prolifer- the protein kinase C and inducing Ca2+ influx, respectively,
ative capacity. We detected transcription factors, PouV, Sox2, bypassing the TCR signaling. PMA and ionomycin-induced
and Nanog, in chicken MSCs but not in skin fibroblasts. PouV proliferation was also suppressed by MSCs (Fig. 4B), suggest-
is a chicken homolog of mammalian Oct4 and like Oct4 has ing that MSCs can modulate signals downstream of protein
CHICKEN MESENCHYMAL STEM CELLS FROM BONE MARROW 1489

A B

C D

E F G
Induction Media Induction Media Induction Media
– + – + – +
PPAR-γ Osteopontin Collagen II

β-Actin β-Actin
aP2

β-Actin

FIG. 3. Multilineage differentiation of chicken mesenchymal stem cells (MSCs) in vitro. (A and B) Adipocyte differentia-
tion. Chicken MSCs were cultured in adipogenic medium. Representative photomicrographs of chicken MSCs are shown
(bright field; A) and stained with the Oil Red O dye. Accumulation of lipids into intracellular vesicles (B). (C) Osteocyte dif-
ferentiation. MSCs were cultured in osteogenic medium. Calcium deposition and mineralization were shown in monolayers
using von Kossa staining. (D) Chondrogenic differentiation. Micromass cultures of chicken MSCs cultured in the presence
of 10 ng/mL TGF-β1 showed a homogenous Alcian blue staining of proteoglycans. (E–G) RT-PCR showing the up-regulation
of the lineage-specific genes in adipogenic-, osteogenic-, and chondrogenic-differentiated cells, respectively. aP2, PPAR-γ,
osteopontin, and collagen II gene expression was induced in differentiated cells.

kinase C and Ca2+ influx and the T-cell receptor complex is derived MSCs expressed transcription factors PouV, Sox2,
not a target for the suppression. and Nanog and differentiated along osteogenic and adipo-
genic lineages when cultured under the appropriate con-
T-cell suppression and NO ditions. Also, the bone marrow-derived MSCs inhibited
mitogen-induced proliferation of splenocytes. Therefore, we
The exact mechanism of MSCs-induced immunosup- conclude that these cells isolated from chicken bone marrow
pression is yet to be defined, but several chemokines and are MSCs and exhibit features similar to those of mamma-
molecules of interest have been identified. NO produced by lian MSCs.
macrophages has been reported to suppress T-cell prolifer- We detected gene expression of PouV in cultured chicken
ation. In a recent study, Sato et al. [32] showed that murine MSCs at passages 2 and 8. PouV is a chicken homolog of Oct4
MSCs T cells co-culture produce NO. This prompted us to in mammals [30]. In mammals, Oct4 is expressed in embry-
examine the production of NO in the chicken MSCs spleno- onic stem cells where it inhibits tissue-specific genes and
cytes co-culture system. Co-culture of chicken MSCs with regulates self-renewal and pluripotency [33]. Recent reports
T cells induced a significant (P < 0.05) and dose-dependent have indicated the expression of Oct4 in MSCs [23,34]. Oct4
production of NO (Fig. 5). NO production was not significant interacts with other embryonic regulators, such as Sox2 and
when MSCs were co-cultured with T cells in the absence of Nanog, which are also involved in the regulation of pluri-
ConA or by ConA-treated MSCs or T cells. High levels of potency and inhibit differentiation [35]. Multilineage differ-
NO were produced in the co-cultures of T cells and MSCs entiation potential of stem cells is strongly correlated with
and this was accompanied by a strong suppression of T-cell the expression of Oct4. Oct4 regulates the expression of sev-
proliferation. eral genes expressed in stem cells, such as fibroblast growth
factor 4 and transcription factors Rex1 and Sox2 [36–39].
Discussion Expression of PouV in MSCs, like chicken embryonic stem
cells may be associated with pluripotency and self-renewal
In the present study, we isolated and expanded adherent of these cells.
cells obtained from chicken bone marrow. The isolated cells In this study, adipogenic differentiation was induced
showed features consistent with MSCs. The bone marrow- with insulin, indomethacin, and dexamethasone, a method
1490 KHATRI, O’BRIEN, AND SHARMA

A 30
140 *
25
120

Nitric Oxide (μM)


% Proliferation

100 20
80 15
60 *
10
40 *
20 5
*
0 0
No MSC 1:10 1:5 MSC Only No MSC 1:10 1:5
B FIG. 5. NO production by splenocytes in the presence of
120
mesenchymal stem cells (MSCs). Splenocytes (2 × 105) were
100 stimulated with ConA (5 μg/mL) in the presence or absence
% Proliferation

80 of the indicated number of MSCs for 48 h. After the incuba-


tion, the supernatant was harvested and examined for NO
60
production by Griess assay. The values are averages ± SD
40 of triplicate determinations. The data are representative of
20 * three separate experiments. *Significantly different from the
MSC Only and No MSC group, P < 0.05.
0
No MSC 1:10

FIG. 4. Mesenchymal stem cells (MSCs) suppress T-cell Several mechanisms of T-cell suppression by MSCs have
proliferation induced by ConA (A) and PMA and ionomycin been proposed including mediation by regulatory T cells,
(B). Splenocytes (2 × 105) were stimulated with ConA (5 μg/ antigen-presenting cells, and immunosuppressive cytokines
mL) or PMA and ionomycin (50 ng/mL and 1 μg/mL, respec- [44–46]. Recently, Sato et al. [32] showed the involvement of
tively) in the presence or absence of the indicated number of NO in the suppression of T cells by murine MSCs. They
MSCs in a 96-well plate for 48 h at 37°C. During the last 5 h showed that NO suppresses T-cell proliferation by inhib-
of incubation, the cells were pulsed with 1 µCi per well of 3H iting Stat-5 phosphorylation. We also detected NO from T
thymidine. Cell-associated radioactivity was harvested and cells and MSCs co-cultures and the extent of T-cell suppres-
is shown relative to that in the absence of MSCs. The values sion correlated with the amount of NO production, suggest-
are averages ± SD of triplicate determinations. The data are ing the important role of NO in immunomodulation. The
representative of three separate experiments. *Significantly physiologic role of NO produced in MSCs T-cell co-culture
different from the No MSC group, P < 0.05. system is unknown. Speculatively, NO might be involved in
the mechanisms by which MSCs in bone marrow protect he-
matopoietic stem cells from T-cell-mediated destruction by
inhibiting T-cell proliferation.
In conclusion, we have demonstrated for the first time
routinely used to stimulate the adipogenic differentiation
that MSCs can be isolated from the bone marrow of hatched
of stem cells. The exact mechanisms of action are not fully
chicken and these cells are capable of in vitro multiplication
known yet; it is believed that the insulin/IGF-1, glucocorti-
and multilineage differentiation, thus making them a suit-
coids, and cAMP signaling pathways mediate the lipid accu-
able model in the field of stem cell research.
mulation in the cells [40]. Differentiation was apparent by
the accumulation of lipid vacuoles within cells, which even-
tually filled the entire cell over a period of time. Cellular dif- Author Disclosure Statement
ferentiation was accompanied by the expression of aP2 and No competing financial interests exist.
PPAR-γ2. PPAR-γ2 is an adipocyte-specific transcription fac-
tor, induced early during the differentiation of adipocytes
References
[41]. It plays an important role in adipocytes differentiation
and also stabilizes the metabolic function of differentiated 1. Jiang Y, BN Jahagirdar, RL Reinhardt, RE Schwartz, CD Keene,
adipocytes [42]. aP2 is also an adipose tissue-specific gene XR Ortiz-Gonzalez, MReyes, T Lenvik, T Lund, M Blackstad, J
and is expressed in the terminal stage of adipocyte differ- Du, S Aldrich, A Lisberg, WC Low, DA Largaespada and CM
entiation [40]. Verfaillie. (2002). Pluripotency of mesenchymal stem cells de-
The osteogenic potential of chicken MSCs was con- rived from adult marrow. Nature 418:41–49.
firmed by demonstration of mineralization in the extracel- 2. Pittenger MF, AM Mackay, SC Beck, RK Jaiswal, R Douglas, JD
Mosca, MA Moorman, DW Simonetti, S Craig and DR Marshak.
lular matrix produced by the differentiated cells, and by the
(1999). Multilineage potential of adult human mesenchymal
expression of osteopontin gene. Treatment of MSCs with
stem cells. Science 284:143–147.
osteogenic induction media resulted in transformation of 3. Pereira RF, KW Halford, MD O’Hara, DB Leeper, BP Sokolov,
cells from elongated fibroblastic to shorter cuboidal cells. In MD Pollard, O Bagasra and DJ Prockop. (1995). Cultured ad-
primary culture, differentiated osteoblasts grew as colonies herent cells from marrow can serve as long-lasting precursor
and formed mineralized nodules, a characteristic of osteo- cells for bone, cartilage, and lung in irradiated mice. Proc Natl
genesis in MSCs of humans [43]. Acad Sci USA 92:4857–4861.
CHICKEN MESENCHYMAL STEM CELLS FROM BONE MARROW 1491

4. Wakitani S, T Saito and AI Caplan. (1995). Myogenic cells de- 23. Izadpanah R, T Joswig, F Tsien, J Dufour, JC Kirijan and BA
rived from rat bone marrow mesenchymal stem cells exposed Bunnell. (2005). Characterization of multipotent mesenchymal
to 5-azacytidine. Muscle Nerve 18:1417–1426. stem cells from the bone marrow of rhesus macaques. Stem
5. Kadiyala S, RG Young, MA Thiede and SP Bruder. (1997). Cells Dev 14:440–451.
Culture expanded canine mesenchymal stem cells pos- 24. Kim IJ, SK You, H Kim, HY Yeh and JM Sharma. (2000).
sess osteochondrogenic potential in vivo and in vitro. Cell Characteristics of bursal T lymphocytes induced by infectious
Transplant 6:125–134. bursal disease virus. J Virol 74:8884–8892.
6. Mosca JD, JK Hendricks, D Buyaner, J Davis-Sproul, LC Chuang, 25. Pertile TL, JM Sharma and MM Walser. (1995). Reovirus infec-
MK Majumdar, R Chopra, F Barry, M Murphy, MA Thiede, tion in chickens primes splenic adherent macrophages to pro-
U Junker, RJ Rigg, SP Forestell, E Bohnlein, R Storb and BM duce nitric oxide in response to T cell-produced factors. Cell
Sandmaier. (2000). Mesenchymal stem cells as vehicles for gene Immunol 164:207–216.
delivery. Clin Orthop Relat Res 379:S71–S90. 26. Nichols J, B Zevnik, K Anastassiadis, H Niwa, D Klewe-
7. Martin DR, NR Cox, TL Hathcock, GP Niemeyer and HJ Baker. Nebenius, I Chambers, H Scholer and A Smith. (1998). Formation
(2002). Isolation and characterization of multipotential mes- of pluripotent stem cells in the mammalian embryo depends on
enchymal stem cells from feline bone marrow. Exp Hematol the POU transcription factor Oct4. Cell 95:379–391.
30:879–886. 27. Chambers I, D Colby, M Robertson, J Nichols, S Lee, S Tweedie
8. Young HE, EM Ceballos, JC Smith, ML Mancini, RP Wright, and A Smith. (2003). Functional expression cloning of Nanog,
BL Ragan, I Bushell and PA Lucas. (1993). Pluripotent mesen- a pluripotency sustaining factor in embryonic stem cells. Cell
chymal stem cells reside within avian connective tissue ma- 113:643–655.
trices. In Vitro Cell Dev Biol Anim 29:723–736. 28. Avilion AA, SK Nicolis, LH Pevny, L Perez, N Vivian and R
9. Young HE, ML Mancini, RP Wright, JC Smith, AC Black, CR Lovell-Badge. (2003). Multipotent cell lineages in early mouse
Reagan Jr. and PA Lucas. (1995). Mesenchymal stem cells re- development depend on SOX2 function. Genes Dev 17:126–140.
side within the connective tissues of many organs. Dev Dyn 29. Hanna LA, RK Foreman, IA Tarasenko, DS Kessler and PA
202:137–144. Labosky. (2002). Requirement for Foxd3 in maintaining pluripo-
10. Iyer SS and M Rojas. (2008). Anti-inflammatory effects of mes- tent cells of the early mouse embryo. Genes Dev 16:2650–2661.
enchymal stem cells: novel concept for future therapies. Expert 30. Lavial F, H Acloque, F Bertocchini, DJ Macleod, S Boast, E
Opin Biol Ther 8:569–581. Bachelard, G Montillet, S Thenot, HM Sang, CD Stern, J Samarut
11. Haynesworth SE, MA Baber and AI Caplan. (1996). Cytokine ex- and B Pain. (2007). The Oct4 homologue PouV and Nanog regu-
pression by human marrow-derived mesenchymal progenitor late pluripotency in chicken embryonic stem cells. Development
cells in vitro: effects of dexamethasone and IL-1 alpha. J Cell 134:3549–3563.
Physiol 166:585–592. 31. Zhao M, SA Amiel, S Ajami, J Jiang, M Rela, N Heaton and GC
12. Majumdar MK, MA Thiede, JD Mosca, M Moorman and SL Huang. (2008). Amelioration of streptozotocin-induced diabetes
Gerson. (1998). Phenotypic and functional comparison of cul- in mice with cells derived from human marrow stromal cells.
tures of marrow-derived mesenchymal stem cells (MSCs) and PLoS ONE 3:e2666.
stromal cells. J Cell Physiol 176:57–66. 32. Sato K, K Ozaki, I Oh, A Meguro, K Hatanaka, T Nagai, K Muroi
13. Awad HA, GP Boivin, MR Dressler, FN Smith, RG Young and and K Ozawa. (2007). Nitric oxide plays a critical role in sup-
DL Butler. (2003). Repair of patellar tendon injuries using a cell– pression of T-cell proliferation by mesenchymal stem cells.
collagen composite. J Orthop Res 21:420–431. Blood 109:228–234.
14. Noel D, F Djouad and C Jorgense. (2002). Regenerative medicine 33. Pan GJ, ZY Chang, HR Scholer and D Pei. (2002). Stem cell pluri-
through mesenchymal stem cells for bone and cartilage repair. potency and transcription factor Oct4. Cell Res 12:321–329.
Curr Opin Investig Drugs 3:1000–1004. 34. Greco SJ, K Liu and P Rameshwar. (2007). Functional similari-
15. Pelttari K, E Steck and W Richter. (2008). The use of mesen- ties among genes regulated by OCT4 in human mesenchymal
chymal stem cells for chondrogenesis. Injury 39:S58–S65. and embryonic stem cells. Stem Cells 25:3143–3154.
16. Pittenger MF and BJ Martin. (2004). Mesenchymal stem cells 35. Boiani M and HR Scholer. (2005). Regulatory networks in embryo-
and their potential as cardiac therapeutics. Circ Res 95:9–20. derived pluripotent stem cells. Nat Rev Mol Cell Biol 6:872–884.
17. Koc ON, SL Gerson, BW Cooper, SM Dyhouse, SE Haynesworth, 36. Ben-Shushan E, JR Thompson, LJ Gudas and Y Bergman. (1998).
AI Caplan and HM Lazarus. (2000). Rapid hematopoietic re- Rex-1, a gene encoding a transcription factor expressed in the
covery after coinfusion of autologous-blood stem cells and cul- early embryo, is regulated via Oct-3/4 and Oct-6 binding to an
ture-expanded marrow mesenchymal stem cells in advanced octamer site and a novel protein, Rox-1, binding to an adjacent
breast cancer patients receiving high-dose chemotherapy. J Clin site. Mol Cell Biol 18:1866–1878.
Oncol 18:307–316. 37. Botquin V, H Hess, G Fuhrmann, C Anastassiadis, MK Gross, G
18. Le Blanc K, I Rasmusson, B Sundberg, C Gotherstrom, M Vriend and HR Scholer. (1998). New POU dimer configuration
Hassan, M Uzunel and O Ringden. (2004). Treatment of severe mediates antagonistic control of an osteopontin preimplanta-
acute graft-versus-host disease with third party haploidentical tion enhancer by Oct-4 and Sox-2. Genes Dev 12:2073–2090.
mesenchymal stem cells. Lancet 363:1439–1441. 38. Niwa H, J Miyazaki and AG Smith. (2000). Quantitative expres-
19. Le Blanc K and O Ringden. (2006). Mesenchymal stem cells: sion of Oct-3/4 defines differentiation, dedifferentiation or self-
properties and role in clinical bone marrow transplantation. renewal of ES cells. Nat Genet 24:372–376.
Curr Opin Immunol 18:586–591. 39. Yuan H, N Corbi, C Basilico and L Dailey. (1995). Developmental-
20. Kassis I, N Grigoriadis, B Gowda-Kurkalli, R Mizrachi-Kol, specific activity of the FGF-4 enhancer requires the synergistic
T Ben-Hur, S Slavin, O Abramsky and D Karussis. (2008). action of Sox2 and Oct-3. Genes Dev 9:2635–2645.
Neuroprotection and immunomodulation with mesenchymal 40. Gregoire FM, CM Smas and Sul HS. (1998). Understanding adi-
stem cells in chronic experimental autoimmune encephalomy- pocyte differentiation. Physiol Rev 78:783–809.
elitis. Arch Neurol 65:753–761. 41. Rosen ED, P Sarraf, AE Troy, G Bradwin, K Moore, DS Milstone,
21. Cooper MD, DA Raymond, RD Peterson, MA South and RA BM Spiegelman and RM Mortensen. (1999). PPAR gamma is re-
Good. (1966). The functions of the thymus system and the bursa quired for the differentiation of adipose tissue in vivo and in
system in the chicken. J Exp Med 123:75–102. vitro. Mol Cell 4:611–617.
22. Nadri S, M Soleimani, RH Hosseni, M Massumi, A Atashi and 42. Lazar MA. (2002). Becoming fat. Genes Dev 16:1–5.
R Izadpanah. (2007). An efficient method for isolation of murine 43. Bruder SP, N Jaiswal and SE Haynesworth. (1997). Growth
bone marrow mesenchymal stem cells. Int J Dev Biol 51:723–729. kinetics, self-renewal, and the osteogenic potential of purified
1492 KHATRI, O’BRIEN, AND SHARMA

human mesenchymal stem cells during extensive subculti- Address correspondence to:
vation and following cryopreservation. J Cell Biochem 64: Dr. Mahesh Khatri
278–294. Department of Veterinary and Biomedical Sciences
44. Aggarwal S and MF Pittenger. (2005). Human mesenchymal University of Minnesota
stem cells modulate allogeneic immune cell responses. Blood
256 Vet. Sci. Building
105:1815–1822.
45. Krampera M, S Glennie, J Dyson, D Scott, R Laylor, E Simpson
1971 Commonwealth Avenue
and F Dazzi. (2003). Bone marrow mesenchymal stem cells in- St. Paul, MN 55108
hibit the response of naive and memory antigen-specific T cells
to their cognate peptide. Blood 101:3722–3729. E-mail: khatr001@umn.edu
46. Rasmusson I, O Ringden, B Sundberg and K Le Blanc. (2003).
Mesenchymal stem cells inhibit the formation of cytotoxic T Received for publication August 6, 2008
lymphocytes, but not activated cytotoxic T lymphocytes or nat- Accepted after revision April 21, 2009
ural killer cells. Transplantation 76:1208–1213. Prepublished on Liebert Instant Online April 21, 2009

Das könnte Ihnen auch gefallen