You are on page 1of 10

Isolation and characterization of mesenchymal stem cells derived from bone

marrow of patients with Parkinson's disease


Author(s): Zhiqing Zhang, Xiaofang Wang, Suping Wang
Source: In Vitro Cellular & Developmental Biology - Animal, 44(5):169-177.
Published By: Society for In Vitro Biology
https://doi.org/10.1007/s11626-008-9093-1
URL: http://www.bioone.org/doi/full/10.1007/s11626-008-9093-1

BioOne (www.bioone.org) is a nonprofit, online aggregation of core research in the biological, ecological,
and environmental sciences. BioOne provides a sustainable online platform for over 170 journals and books
published by nonprofit societies, associations, museums, institutions, and presses.
Your use of this PDF, the BioOne Web site, and all posted and associated content indicates your acceptance of
BioOne’s Terms of Use, available at www.bioone.org/page/terms_of_use.
Usage of BioOne content is strictly limited to personal, educational, and non-commercial use. Commercial
inquiries or rights and permissions requests should be directed to the individual publisher as copyright holder.

BioOne sees sustainable scholarly publishing as an inherently collaborative enterprise connecting authors, nonprofit publishers, academic institutions,
research libraries, and research funders in the common goal of maximizing access to critical research.
In Vitro Cell.Dev.Biol.—Animal (2008) 44:169–177
DOI 10.1007/s11626-008-9093-1

Isolation and characterization of mesenchymal stem cells


derived from bone marrow of patients
with Parkinson’s disease
Zhiqing Zhang & Xiaofang Wang & Suping Wang

Received: 10 December 2007 / Accepted: 29 February 2008 / Published online: 10 April 2008 / Editor: J. Denry Sato
# The Society for In Vitro Biology 2008

Abstract Mesenchymal stem cells (MSCs) are capable of Introduction


self-renewing and differentiating into multiple tissues; they
are expected to become a source of cells for regenerative Bone marrow stroma contains multipotential mesenchymal
therapy. Compared to allogeneic MSCs, autologous MSCs stem cells (MSCs) capable of supporting hematopoietic
from patients needing cell-based therapy may be an ideal stem cell growth, self-renewing, and differentiating into
alternative stem cell source. However, characterizations of various tissues of mesenchymal lineages, such as bone,
MSCs from a disease state remains extremely limited. cartilage, tendon, muscle, and fat under specific conditions
Therefore, we have isolated and characterized MSCs from (Horwitz et al. 1999; Makino et al. 1999; Pittenger et al.
Parkinson’s disease (PD) patients and compared them with 1999; Jiang et al. 2002; Zhao et al. 2002; Kasten et al.
MSCs derived from normal adult bone marrow. Our results 2006). Moreover, MSCs can be obtained from adult
show that PD-derived MSCs are similar to normal MSCs in organisms and can be expanded more than 1 billion fold
phenotype, morphology, and multidifferentiation capacity. in ideal culture without the loss of their characteristics of
Moreover, PD-derived MSCs are capable of differentiating stem cells. Due to these abilities, MSCs are of great
into neurons in a specific medium with up to 30% having potential in the context of tissue engineering and cell-based
the characteristics of dopamine cells. At last, PD-derived therapy (Minguell et al. 2000; Le Blanc et al. 2004).
MSCs could inhibit T-lymphocyte proliferation induced by Previous studies have indicated that both mice and
mitogens. These findings indicate that MSCs derived from human MSCs could be induced to differentiate into neurons
PD patients’ bone marrow may be a promising cell type for in vitro by β-mercaptoethanol or dimethylsulfoxide, and
cellular therapy and somatic gene therapy applications. butylated hydroxyanisol or in vivo by engraftment (Mezey
et al. 2000; Sanchez-Ramos et al. 2000; Woodbury et al.
Keywords Mesenchymal stem cells . Bone marrow . 2000; Brazelton et al. 2000; Zhang et al. 2006). Kopen et al.
Parkinson’s disease . Immunosuppression (1999) injected MSCs into the lateral ventricle of neonatal
mice and found that transplanted MSCs could differentiate
into neurons when exposed to the brain microenvironment,
suggesting that MSCs are potentially useful as vectors for
treating a variety of central nervous system disorders.
Recently, studies demonstrated that MSCs could be induced
Z. Zhang (*) : S. Wang
into tyrosine hydroxylase-positive and dopamine-producing
Department of Neurology, Dalian Central Hospital,
Dalian 116033, People’s Republic of China neuronal cells in vitro. Transplantation of these induced
e-mail: zhangzhiqing1972@yahoo.com.cn cells showed improvement in apomorphine-induced rota-
tional behavior and adjusting step and paw-reaching tests
X. Wang
following intrastriatal implantation in a 6-hydroxy dopa-
Department of Hematology and Oncology,
the Oncology Hospital of Tianjin Medical University, mine rat model of Parkinson’s disease (PD; Dezawa et al.
Tianjin, People’s Republic of China 2004). These findings demonstrated that the ability to
170 ZHANG ET AL.

transdifferentiate MSCs into special neurons could have a CD14 micromagnetic beads (Miltenyi Biotec, Auburn,
substantial potential for treating neurodegenerative disorders. CA), and CD14-negative cells were replated.
Compare to allogeneic MSCs, autologous MSCs are
more suitable for clinical applications, especially for those Quantification of DNA content and cell growth patterns.
needing cell-based therapy. However, previous studies During the log phase of growth, the expanded MSCs were
only demonstrated the characteristics of MSCs derived trypsinized, permeabilized with 70% ethanol for 20 min at
from health donors, and the definitive characteristics of 4° C, washed with phosphate-buffered saline (PBS), then
MSCs derived from disease states remain extremely treated with 40 μg/mL RNAse A for 20 min at 37° C and in
limited. In this study, we have made an attempt to isolate 10 μg/mL propidium iodine for 5 min at room temperature.
and culture autologous MSCs from PD patients’ bone Deoxyribonucleic acid (DNA) content was analyzed in
marrow and examine their biology characteristics. Our FACScan flow cytometer (Becton Dickinson, Franklin
data showed that autologous MSCs derived from PD Lakes, NJ) using the Cellquest software. To study the
patients display a fibroblast-like morphology, express growth pattern of the expanded MSCs, cells were seeded at
mesenchymal phenotype, and are able to differentiate into a concentration of 2×103 cells per well in a 24-well plate.
adipocytes, osteoblasts, and neural cells in vitro. More- The adherent cells were trypsinized and counted by the
over, PD-derived MSCs are capable of differentiating into 0.4% trypan blue exclusion method (dead cells would be
neurons under specific conditions with up to 30% having stained by trypan blue and thus excluded) at different time
the characteristics of dopamine cells. At last, PD-derived points. Moreover, doubling time of the MSCs for passages
MSCs possessed hematopoietic supportive ability and 3–15 was determined by seeding 1×105 cells into 25-cm2
could inhibit T lymphocyte proliferation induced by T-flasks, which were fed with expanded medium every 2 d
mitogens. and then trypsinized and counted by the 0.4% trypan blue
exclusion method after 4 d. Mean doubling time was
calculated from days 0 to 4 for four different origin MSCs.
Materials and Methods
FACS analysis. For immunophenotype analysis, MSCs
Isolation of MSCs from PD patients. Eighteen patients with were washed with PBS containing 0.5% bovine serum
PD (11 men, 7 women) and eight (five men, three women) albumin (Sigma) and incubated with primary antibodies
normal healthy volunteers were investigated in this study. (10–20 ng/mL) for 30 min at 4° C. For intracellular
The mean age of the PD patients was 60.7 years (range 48– antigens detection, cells were fixed in 2% paraformalde-
69), and the mean age of the controls was 58.4 years (range hyde for 15 min at 4° C and permeabilized with 0.1%
52–64). None of the patients had received any antiparkin- saponin (Sigma) for 1 h at room temperature. Primary
sonian medication. None of the control subjects had a antibodies included mAb against CD11a, CD11b, CD14,
history of neurologic or psychiatric diseases. After receiv- CD29, CD31, CD34, CD44, CD45, CD105, CD106, GlyA,
ing informed consent from the patients according to the and HLA-DR (BD Biosciences Pharmingen, San Diego,
academic guidelines on the use of human subjects in CA). We used same-species, same-isotype irrelevant anti-
research, human bone marrow was obtained from PD body as the negative control. Then, cells were washed and
patients and healthy donors under the protocol approved incubated with fluorescein isothiocyanate (FITC)-conjugat-
by the institutional review board of the Dalian Central ed secondary antibodies for 30 min at 4° C. Cell analysis
Hospital. Bone marrow samples were taken from the iliac was performed with a FACSCalibur system using CellQuest
crest of normal adults or PD patients. Mononuclear cells software. Each measurement contained 10,000 cells.
were separated by a Ficoll–Paque gradient centrifugation
(specific gravity 1.077 g/mL; Sigma Diagnostics, St Louis, Karyotyping analysis. MSCs derived from PD or normal
MO) and cultured in expansion medium at 37° C with 5% adults were analysis for karyotype. All cells in the log
CO2 in a fully humidified atmosphere. The expansion phase of growth were processed by 0.1 μM cochineals for
medium contained 60% Dulbecco’s modified Eagle’s 6 h at 37° C and then digested with 0.25% trypsin–EDTA
medium (DMEM)/F-12 (Gibco Life Technologies, Paisley, and subjected to a 15 min colcemid (Sigma) incubation
UK), 40% MCDB-201 (Sigma), 10% fetal calf serum (FCS; followed by lysis with 0.075 mM KCl and fixation in
Gibco), 100 U/mL penicillin, and 1,000 U/mL streptomycin methanol/acetic acid (3:1). Metaphases were analyzed after
(Gibco). After culture for 2 d, the culture medium was QFQ or GTG banding.
replaced, and nonadherent cells were removed. Once cells
were more than 80% confluent, they were detached with Transmission electron microscope analysis. Once expand-
0.25% trypsin–ethylenediamine tetraacetic acid (EDTA; ed MSCs were more than 90% confluent, they were
Sigma), and then CD14-positive cells were depleted using detached with 0.25% trypsin–0.04%EDTA and centrifuged
FUNCTION OF AUTOLOGOUS MESENCHYMAL STEM CELL 171

at 1,200 rpm for 5 min, and then the cells blocks were fixed 2004). Cells were washed twice with a low K+ solution
in 6% glutaraldehyde and 1% OsO4. After dehydration in and incubated in the low K+ solution for 2 min, and then
acetone, cells blocks were embedded in GP-812 resin and the medium was replaced with a high K+ solution for 5 min.
cut into ultrathin sections. Sections were double-stained Concentration of dopamine was determined by high-
with uranyl acetate and lead citrate before observation performance liquid chromatography (HPLC) using a re-
under an electron microscope. verse-phase column and an electrochemical detector system
(Eicom, Kyoto, Japan; Dezawa et al. 2004).
Multilineage differentiation of MSCs. For osteoblasts dif-
ferentiations, MSCs were cultured at 2–3×104/cm2 in Mixed leukocyte reaction. MSCs were irradiated (30 Gy)
DMEM with 10 mM β-glycerophosphate, 10−7 M dexa- and plated in triplicates onto 96-well plates at 1×104/mL in
methasone, and 0.2 mM ascorbic acid. After 21 d, 100 μL complete media. Allogeneic CD2+ T cells purified
differentiation to osteoblasts was shown by Von Kossa from peripheral blood mononuclear cells by using the
stain for calcium phosphate and reverse transcriptase (RT)- MACS CD4 isolation kit, resuspended at 1×105/mL, were
polymerase chain reaction (PCR) for osteopontin; For added to wells containing or lacking MSCs in the presence
adipocyte differentiations, expanded clonal MSCs were of the mitogen phytohemagglutinin (PHA; 2.5 μg/mL). The
cultured at 2–3×104/cm2 in DMEM with 10% FCS, 10−6 M MSCs to CD4+ T cells ratio was 1:10. Cocultures without
dexamethasone, 50 μg/mL ascorbic acid, and 100 μg/mL 1- PHA were used as controls. The culture was continued, and
methyl -3- isobutyl-xanthine (Sigma). After 14 d, oil-red 3
H-thymidine (1 μCi [0.037 MBq]) was added 18 h before
staining and RT-PCR for lipoprotein lipase showed adipo- the end of the 120-h culture. The T cell proliferation was
cyte differentiation; for chondrogenic differentiation, 1×106 represented as the incorporated radioactivity in counts per
MSCs were cultured in 1 mL serum-free expansion medium minute (cpm) and shown as means±SDs of triplicate
with 100 ng/mL transforming growth factor 1, in the tip of values.
a 15-mL conical tube to allow aggregation of the cells in
the micromass suspension culture. The development of Reverse transcriptase polymerase chain reaction. RT-PCR
chondroblasts was followed by staining the micromasses for lipoprotein lipase, osteopontin, transcription factors
with toluidine blue, which stains the proteoglycan extracel- Nurr-1, Lmx1b, Ptx3, and β2-microglobulin was per-
lular matrix (ECM), and with antibodies against type II formed. Total ribonucleic acid (RNA) was prepared with
collagen. For neural differentiation, MSCs were cultured in the use of Trizol reagents in accordance with the protocol
a 24-well plate at 2×104 cells per well in α-modified recommended by the manufacturer and visualized by means
minimum essential medium (αMEM) with 10% FCS, of UV spectrophotometry. We synthesized first-strand
10 ng/mL basic fibroblast growth factor (bFGF; Peprotech, complementary DNA (cDNA) from 4 μg of total RNA
Rocky Hill, NJ), 50 ng/mL brain-derived neurotrophic primed with oligo-dT primer using the First-Strand cDNA
factor (Peprotech), and 50 ng/mL of Nerve growth factor Syn-thesis Kit (Gibco). We amplified 1/50 of the RT
(Takara Shuzo, Tokyo, Japan). The differentiated cells were mixture with the use of the GeneAmp PCR System 2400
observed by phase-contrast microscopy and detected by thermal cycler (Perkin-Elmer, Boston, MA) in a total
fluorescence-activated cell sorting (FACS), immunocyto- reaction volume of 20 μL containing 10 pmol of each
chemistry, and Western blotting for neuron-specific markers primer, 200 μmol/L of deoxyribinucleoside triphosphates,
microtubule-associated protein 2 (MAP-2) and neurofila- and 2 U of Taq polymerase (Gibco). Primer sequences and
ment (NF), astrocyto-specific marker, glial fibrillary acidic precise conditions are described elsewhere (Seta et al. 1999;
protein (GFAP), and oligodendrocytic-specific marker Zheng et al. 2000; Fang et al. 2005). The reaction products
galactocerebroside (GalC; Sanchez-Ramos et al. 2000). were resolved by electrophoresis on a 1.5% agarose gel and
visualized with ethidium bromide.
Dopaminergic neuron differentiation of MSCs. For dopa-
minergic neuron differentiation, MSCs were cultured in a Immunofluorescence and Western blotting. For immunoflu-
24-well plate at 2×104cells per well in αMEM with 10% orescence, neural-like cells were fixed with 4% parafor-
FCS, 10 ng/mL bFGF (Peprotech), and 50 ng/mL of GDNF maldehyde at room temperature, permeabilized them with
(Peprotech). The differentiated cells were examined by the use of 0.1% Triton X-100 in PBS for 10 min, and
FACS, immunofluorescence, RT-PCR, and Western blotting treated them with 5% FCS in PBS for 30 min at room
for dopaminergic neuron-specific marker tyrosine hydrox- temperature. Cells then were incubated for 1 h with diluted
ylase (TH) and transcription factors Nurr-1, Lmx1b, and primary antibodies at room temperature. Mouse anti-human
Ptx3 (Sakurada et al. 1999; Dezawa et al. 2004). Further- GFAP (BD Pharmingen; 1:1,000 dilution), GalC (Serotec,
more, differentiated cells were examined by dopamine Raleigh, NC; 1:500 dilution), and NF (Santa Cruz Biotech-
release assay as described previously (Dezawa et al. nology, Santa Cruz, CA; 1:100 dilution) were used. After
172 ZHANG ET AL.

incubated with these antibodies for 1 h at room tempera-


ture, samples were washed three times with PBS and then
incubated for 30 min at room temperature with anti-mouse
IgG/FITC (Sigma). Next, the slides were scrutinized, field
by field, with the use of fluorescence microscopy (Olympus;
Olympus Optical, Tokyo, Japan).
For Western blotting, equivalent amount of protein
lysates, obtained from MSCs or differentiated progeny,
were loaded per lane. After sodium dodecyl sulfate
polyacrylamide gel electrophoresis, proteins were electro-
phoretically transferred onto nitrocellulose membrane
(Amersham Pharmacia Biotech; Uppsala, Sweden). After
blocking, blots were incubated with mouse polyclonal
antibodies against GalC (Serotec; 1:500 dilution), GFAP
(BD Pharmingen; 1:1,000 dilution), MAP-2 (Dako, Carpin-
teria, CA; 1:100 dilution), NF (Santa Cruz Biotechnology;
1:100 dilution), and TH (1:200 dilution) for one night at 4° C
temperature. Expression of the β-actin was used as an
internal control. Immunodetection using the enhanced
chemiluminescence method (ECL kit; Amersham, Piscat-
away, NJ) was performed according to the manufacturer’s
instructions. Figure 1. Morphology and growth characteristic of PD MSCs. A. PD
MSCs were cultured at 2×103 cells per well in a 24-well plate. The
number of viable cells was measured by counting trypan blue-negative
cells. The population doubling time was about 50 h. B. Doubling time
of MSCs derived from normal and PD MSCs with successive passage.
Result Results are expressed as mean±SEM. Normal adult, diamonds, n=4;
PD, squares, n=4.
Biological characteristics of undifferentiated PD MSCs. In
this study, MSCs were obtained from the bone marrow of MSCs derived from PD and normal adults showed a normal
PD patients (n=18). We compare their biological character- karyotype.
istics with MSCs derived from bone marrow of normal
adults (n=8). The biological characteristics of MSCs were Ultrastructure of undifferentiated PD MSCs. Under trans-
investigated and compared over a 20-wk period. Our results mission electron, the nucleus of the MSCs contains one or
show that MSCs derived from PD and normal patients two small nucleoli surrounded by nucleolar-associated
displayed fibroblast-like morphology under phase-contrast chromatin. The cytoplasm contains a relatively small
microscopy. Two weeks later, almost all obtained MSCs (16 quantity of organelles, such as the endoplasmic reticulum,
of 18 for PD and eight of eight for normal adult) formed ribosomes, mitochondria, and Golgi complex. There are no
colony-forming unit fibroblast colonies (CFU-F). The granules in the cytoplasm. Moreover, normal and PD MSCs
frequency of CFU-F was similar among MSCs derived have a similar ultrastructure.
from PD and normal patients (4±1.89 and 5±2.14 CFU-F
per 106 bone marrow mononuclear cells for PD and normal Multiple differentiation of PD MSCs. To induce adipocyte
MSCs, respectively). A typical growth curve is shown in differentiation, there were lipid-laden cells that appeared
Fig. 1A. The doubling time of PD and normal-origin MSCs after 5 d. After 2 wk, more than 60% of MSCs
was similar (52.3±6.5 h for PD, 51.9±10.3 h for normal differentiated into adipocytes that stained with Oil-red O
adult, respectively). The doubling time of MSCs remained (Fig. 2A, C). Using RT-PCR, we also found that these cells
unchanged during culture expand for at least 20 passages expressed lipoprotein lipase, an adipocyte special gene
(Fig. 1B). On average, 91.34% cells were at G0/G1, 2.71% (Fig. 2E). At exposure of MSCs to the osteogenic medium,
at G2/M, and 5.95% at S phases of the cell cycle. The the cells formed aggregates, and there were calcium deposit
immunophenotype of PD MSCs was analyzed by FACS. appears after 3 wk. To confirm osteogenic differentiation,
PD MSCs expressed CD106, CD105, CD29, and CD44; Von Kossa staining for ECM with one osteogenic-specific
they did not express CD11a, CD11b, CD14, CD31, CD34, marker was performed, and we identified that calcification
CD45, GlyA, and HLA-DR (Table 1). This phenotype of the ECM was observed in induced MSCs (Fig. 2B, D).
remained unchanged for more than 20 passages. Moreover, Moreover, RT-PCR analysis also showed another osteogenic-
FUNCTION OF AUTOLOGOUS MESENCHYMAL STEM CELL 173

Table 1. Immunophenotypes of cultured MSCs data showed that induced cells released 2.14 pmol/106 cells
Antigen PD MSCs (%) Normal adult MSCs (%) of dopamine to the culture media in response to high K+-
depolarizing stimuli, whereas dopamine release from
CD11a −0.8 (0.3–1.6) −0.9 (0.3–1.8) undifferentiated MSCs was under the detection level.
CD11b −1.2 (0.5–1.4) −1.1 (0.4–1.5)
CD14 −0.9 (0.4–1.5) −0.8 (0.7–1.2)
Inhibition of T lymphocyte proliferation. Previous reports
CD29 +99.7 (98.2–100) +98.1 (97.3–99.7)
reported that MSCs are capable of suppressing the
CD31 −0.7(0.3–1.7) −1.7 (0.8–2.5)
CD34 −1.3 (0.3–2.1) −0.7 (0.2–0.9) proliferation of T lymphocytes stimulated in a mixed-
CD44 +97.3 (96.2–98.4) +94.8 (93.2–99.4) lymphocyte culture. Therefore, the effect of MSCs on T cell
CD45 −1.0 (0.8–1.4) −0.4 (0.1–0.7) proliferation was evaluated by mixed-leukocyte reactions in
CD105 +96.4 (95.2–98.7) +95.8 (94.7–99.1) the presence of PHA. Our data showed that T lymphocyte
CD106 +94.9 (93.2–99.8) +97.2 (96.6–98.2) proliferation induced by PHA was markedly reduced in the
HLA-DR −1.2 (0.7–2.2) −0.9 (0.7–1.8) presence of PD MSCs or normal adult MSCs. The addition
Glay A −1.4 (0.9–1.8) −0.7 (0.4–1.3)
of MSCs to the mixed T lymphocyte culture could reduce T
− indicated no cells were positively stained; + indicated cells were lymphocyte proliferation by more than 70% (P≤0.01). The
positively stained. ability of inhibition of PHA-stimulated T cell proliferation
was similar between PD and normal adult bone marrow
specific gene, osteopontin, in the cells after osteogenic MSCs (Fig. 4).
induction (Fig. 2E). To induce chondrogenic differentiation,
after 2 wk, small aggregates of cartilage formed in the
bottom of the tubes were stained positive with toluidine blue
(Fig. 2F, G). Moreover, expression of type II collagen was Discussion
detected from day 5, and type II collagen staining could be
detected throughout the micromass (Fig. 2H). Exposure of PD is a common neurodegenerative disorder of the nerve
MSCs to the induced medium resulted in the expected system. The critical pathological change underlying the
changes in morphology as responsive cells assumed forms condition is the loss of the dopaminergic nigrostriatal
typical of nerve-like cells after 7 d. After 14 d, about 40% of pathway with the formation of α-synuclein-positive Lewy
the induced cells exhibited typical morphology of a nerve- bodies. Tremor and slow movements develop only after
like cell. Induced MSCs were assessed for the expression of about 70% of vulnerable dopaminergic neurons in the
neuronal-specific markers (NF) and MAP-2, in addition to substantia nigra has already died (Quinn 1997; Conway and
GFAP and GalC, as markers of astrocytes and oligoden- Harper 1998; Dauer and Przedborski 2003). Current
drocytes, respectively. FACS showed that 40% of induced therapeutic strategies for PD focus primarily on reducing
cells expressed NF. Immunofluorescence and Western the severity of its symptoms using dopaminergic drugs.
blotting confirmed the expression of NF and MAP-2 in Unfortunately, with time, these therapies fail and produce
induced cells. In contrast to NF or MAP-2, no expression of their own unique side effect profile, and this, coupled with
GalC or GFAP was noted, indicating that MSCs cells did not the more diffuse pathological and clinical findings in
differentiate into oligodendrocytes or astrocytes. (Fig. 2I, J) advancing disease, has led to a search for more effective
therapies (Dunnett and Bjorklund 1999; Jankovic 2002;
Dopaminergic neuron differentiation. After cultured in a Ahlskog 2003). More recently, cell-based therapy has
medium containing GDNF for 2 wk, more than 40% of emerged as a promising approach for restoration of function
MSCs differentiated into neuron-like cells that were in neurodegenerative diseases, in particular PD and Hun-
positive for NF. We performed immunocytochemical tington’s disease (Kawasaki et al. 2000; Perrier et al. 2004).
examination of transmitters and related proteins in those Previous studies demonstrated that embryonic stem cells
cells and found that induced cells were immunopositive for (ESCs), obtained from the early stages of embryonic
TH, one of the dopaminergic neuron-specific markers development, and neural stem cells (NSCs), obtained from
(Fig. 3A), and FACS showed that about 30% cells the developing brain, have been proposed as candidates
expressed TH. Western blot analysis further confirmed for transplantation therapy in neurodegenerative diseases
these results (Fig. 3B). Moreover, induced cells also express (Barberi et al. 2003; Park et al. 2004). However, the
the transcription factors of Nurr-1, Lmx1b, and Ptx3, which practical use of ESCs or NSCs was limited due to potential
are the transcription factors that have roles in the problems of cell regulation and ethical considerations.
differentiation of midbrain precursors into dopaminergic Recently, several studies demonstrated that MSCs were of
neurons (Quinn 1997; Fig. 3C). The amount of dopamine great therapeutic potential against neurological diseases
released upon depolarization was measured by HPLC. Our because of reasons as follows: Firstly, they can be readily
174 ZHANG ET AL.

Figure 2. MSCs differentiate into adipocyte, osteoblast, chondroblast, control. PD MSCs (F) and normal-derived
Figure 2. MSCs differentiate into adipocyte, osteoblast, chondroblast, and neuroectoderm in vitro. PD MSCs (A)MSCs (G) were
and normal bonecultured for
marrow-
and neuroectoderm in vitro. PD MSCs (A) and normal bone marrow-
derived MSCs (C) were cultured for 5 d in an induced medium; lipid-laden 2cells
wk appear
in the induced medium
in adipogenic and stained
medium, with adipocyte
and then, toluidine blue to identify
differentiation
derived MSCs (C) were cultured for 5 d in an induced medium; lipid-laden proteoglycan ECM. Moreover, the for
presence
was shown by Oil-red O staining. PD MSCs (B) and normal bone marrow-derived MSCs (D) were cultured 3 wk inofantype II collagen
osteogenic was
medium,
cells appear in adipogenic medium, and then, adipocyte differentiation was confirmed by Western blotfor
(H;lipoprotein
lane 1, induced PDosteopontin
MSCs; lane 2,
and osteoblast differentiation was indicated by Von Kossa staining for calcium phosphate. (E) RT-PCR lipase and gene.
shown by Oil-red O staining. PD MSCs (B) and normal bone marrow- uninduced PD MSCs; lane
The expression of lipoprotein lipase was detected in induced adipocytes, and the osteocalcin gene was3, detected
induced in normal-derived MSCs; (lane
induced osteoblasts lane 3,
4,
derived MSCs (D) were cultured for 3 wk in an osteogenic medium, and uninduced normal-derived MSCs; bottom, β-actin).
induced normal derived MSCs; lane 4, induced PD MSCs), and there was no expression in uninduced cells (lane 1, uninduced normal derived I After 7 d cultured in
osteoblast differentiation was indicated by Von Kossa staining for calcium
MSCs; lane 2, uninduced PD MSCs). β2-Microglobulin was performed asthe neurogenic
a control. medium,
PD MSCs (F)induced cells were stained
and normal-derived MSCspositively
(G) werefor NF. J
cultured
phosphate. (E) RT-PCR for lipoprotein lipase and osteopontin gene. The Neuronal differentiation was detected by Western blotting with monoclo-
for 2 wk in the induced medium and stained with toluidine blue to identify proteoglycan ECM. Moreover, the presence of type II collagen was
expression of lipoprotein lipase was detected in induced adipocytes, and nalPD
antibodies against NF, MAP2, GFAP, and GalC
confirmed by Western blot (H; lane 1, induced PD MSCs; lane 2, uninduced MSCs; lane 3, induced normal-derived MSCs;(lane lane1,4,induced
uninducedPD
the osteocalcin gene was detected in induced osteoblasts (lane 3, induced MSCs; lane 2, uninduced PD MSCs; lane 3, induced normal-derived
normal-derived MSCs; bottom, β-actin). I After 7 d cultured in the neurogenic medium, induced cells were stained positively for NF. J Neuronal
normal derived MSCs; lane 4, induced PD MSCs), and there was no
differentiation was detected by Western blotting with monoclonal antibodies MSCs; laneNF,
against 4, MAP2,
uninduced
GFAP, normal-derived
and GalC (lane MSCs; bottom,PDβ-actin).
1, induced MSCs;
expression in uninduced cells (lane 1, uninduced normal derived MSCs; Original magnifications: A, B, C, D, F, G, and I: 100×. Representative
lane 2, uninduced PD MSCs; lane 3, induced normal-derived MSCs; lane 4, uninduced normal-derived MSCs; bottom, β-actin). Original
lane 2, uninduced PD MSCs). β2-Microglobulin was performed as a example of six experiments.
FUNCTION OF AUTOLOGOUS MESENCHYMAL STEM CELL 175

Figure
Figure 3.3. PD
PD MSCs
MSCs differentiate
differentiateinto
intodopaminergic
dopaminergicneuron
neurondifferentiation.
differen- A. Afterderived
normal cultured in medium
MSCs; bottom,containing
β-actin).GDNF forcultured
C. After 2 wk, induced cells
in medium
were stained
tiation. positively
A. After for TH.
cultured B. Dopaminergic
in medium neuron
containing GDNF differentiation
for 2 wk, wascontaining
detected by
GDNFWestern
for 2blotting with monoclonal
wk, induced antibodies
cells also express against TH.
the transcription
(lane 1, cells
induced uninduced PD MSCs;
were stained lane 2,
positively forinduced PD MSCs; lane
TH. B. Dopaminergic 3, uninduced
neuron factorsnormal derived
of Nurr-1, Lmx1b,MSCs;and lane
Ptx3 4, induced
(lane normal derived
1, uninduced MSCs;
normal derived
bottom, β-actin).
differentiation wasC. After cultured
detected in medium
by Western containing
blotting with GDNF for 2 wk,MSCs;
monoclonal inducedlane
cells2,also express PD
uninduced the MSCs;
transcription
lane 3,factors of Nurr-1,
induced Lmx1b,
normal-derived
and Ptx3 (lane
antibodies 1, uninduced
against normal
TH. (lane 1, derived
uninduced PDMSCs;
MSCs;lane
lane2,2,uninduced
induced PD MSCs;
MSCs; lane
lane 3, induced
4, induced PD normal-derived
MSCs; bottom, β MSCs; lane 4, induced PD
2-microglobulin). Original
MSCs;
PD MSCs; laneβ3,2-microglobulin).
bottom, uninduced normal Original magnification:
derived MSCs; lane A: 100×. Representative
4, induced example
magnification: of sixRepresentative
A: 100×. experiments. example of six experiments.

obtained through a well-established clinical procedure; last, we test the differentiation ability of different passage
secondly, they are easy to isolate and expand for auto- PD MSCs. They had the ability to differentiate into, at least,
transplantation with no risk of rejection; lastly, they are adipocytes, osteoblasts, chondroblasts, and neural cells in
capable of self-renewal and multiple differentiations (Deans vitro, and the differentiation ability of the fifth, tenth, and
and Moseley 2000; Koc and Lazarus 2001; Reyes et al. 15th passage PD MSCs had no difference. Previous studies
2001). had shown that stem cells possessed the capacities of self-
Although both allogeneic and autologous MSCs possess renewal and multidifferentiation, so our results demonstrate
the clinical therapeutic potential, allogeneic MSCs must that PD MSCs meet the criteria for stem cells.
face limitations inherent, such as histocompatibility, inad-
equate tissue supply, therapeutic scale, and ethical con-
cerns. Therefore, autologous MSCs from patients needing
cell-based therapy may be an ideal alternative stem cell
source and are more suitable for clinical applications. In
this study, we investigated the characteristics of MSCs
derived from patients with PD and compared them with
MSCs derived from normal adult bone marrow. Our data
showed that PD MSCs displayed fibroblast-like morphol-
ogy and could withstand continuous passage for at least 20
times. The morphology and phenotype of MSCs remained
unchanged with cell passage, implying that we could obtain
sufficient cell counts for cell-based therapy. PD MSCs
expressed several cell surface antigens commonly found on
so-called MSCs, such as SH2 (CD105), CD44, CD29, and Figure 4. Inhibitory effect of PD MSCs on T-lymphocyte prolifera-
tion. Allogeneic CD2+ T cells were incubated for 5 d with MSCs
CD106. Furthermore, the flow cytometer showed that about
derived from PD or normal adults in the presence of the mitogen PHA.
91% MSCs were in the G0/G1 phase, indicating that the Data are expressed as mean±SD of triplicate values. Asterisk, P≤0.05.
most of MSCs were in the undifferentiated early state. At Representative example of four experiments.
176 ZHANG ET AL.

The multidifferentiation capacity of MSCs has stimulat- derived MSCs are capable of differentiating into neurons
ed research aimed at the selective generation of specific in tissue culture with up to 30% having the characteristics
cells types for regeneration medicine. Previous studies had of dopamine cells. At last, PD-derived MSCs could inhibit
shown that the coculture of human ESCs on stromal feeder T lymphocyte proliferation induced by mitogens. There-
cells yields highly efficient differentiation into dopamine fore, the successful isolation of MSCs from PD patients’
neuron (Kawasaki et al. 2000; Lee et al. 2000). More bone marrow and the examination of their function and
recently, Dezawa et al. (2004) reported that both rat and natural characteristics may lead to new therapeutic
human MSCs have the capability under specific conditions approaches for this disease.
of differentiating into dopamine neurons and that induced
cells may have properties useful in the treatment of PD. Acknowledgment The authors thank all staff of department of
Although dopamine neurons can be obtained form ESCs or Neurology of Dalian People Hospital for the donation and collection
allogeneic MSCs, the inherent defects limit the application of bone marrow samples.
of ESCs or allogeneic MSCs. Compared to ESCs or
allogeneic MSCs, autologous MSCs from patients needing
cell-based therapy may be an ideal alternative stem cell References
source and are more suitable for clinical applications.
Similar to previous studies, our experiments had also Ahlskog J. E. Slowing Parkinson’s disease progression. Neurology 60:
demonstrated that autologous MSCs derived from PD 381–389; 2003.
Barberi T.; Klivenyi P.; Calingasan N. Y.; Lee H.; Kawamata H.;
patients could selectively differentiate into dopaminergic Loonam K. et al. Neural subtype specification of fertilization and
neurons under an induced medium used before. We found nuclear transfer embryonic stem cells and application in
that about 30% induced cells were immunopositive for TH, parkinsonian mice. Nat. Biotechnol. 21: 1200–1207; 2003.
the rate-limiting enzyme in the synthesis of dopamine. In Bartholomew A.; Sturgeon C.; Siatskas M.; Ferrer K.; McIntosh K.;
Patil S. et al. Mesenchymal stem cells suppress lymphocyte
addition to TH expression, induced cells also express the
proliferation in vitro and prolong skin graft survival in vivo. Exp.
transcription factors of Nurr-1, Lmx1b, and Ptx3, which are Hematol. 30: 42–48; 2002.
the transcription factors that have roles in the differentiation Brazelton T. R.; Rossi F. M.; Keshet G. I.; Blau H. M; From marrow
of midbrain precursors into dopaminergic neurons (Sakurada to brain: expression of neuronal phenotypes in adult mice.
Science 290: 1775–1779; 2000.
et al. 1999). Moreover, induced cells could produce
Conway K. A.; Harper J. D. Lansbury PTAccelerated in vitro fibril
dopamine, suggesting the efficient derivation of dopamine formation by a mutant alpha-synuclein linked to early-onset
neurons from PD MSCs. Parkinson disease. Nat. Med. 4: 1318–1320; 1998.
Previous reports reported that MSCs are capable of Dauer W.; Przedborski S. Parkinson’s disease: mechanisms and
models. Neuron 39: 889–909; 2003.
suppressing the proliferation of T lymphocytes stimulated
Deans R. J.; Moseley A. B. Mesenchymal stem cells: biology and
in a mixed-lymphocyte culture or by incubation with potential clinical uses. Exp. Hematol. 28: 875–84; 2000.
mitogens (Di Nicola et al. 2002; Krampera et al. 2003; Dezawa M.; Kanno H.; Hoshino M.; Cho H.; Matsumoto N.; Itokazu
Tse et al. 2003). The immunosuppressive ability of MSCs Y. et al. Specific induction of neuronal cells from bone marrow
stromal cells and application for autologous transplantation. J.
has a substantial impact in transplantation and may reduce
Clin. Invest. 113: 1701–1710; 2004.
the allogeneic immune response. Batholomew et al. (2002) Di Nicola M.; Carlo-Stella C.; Magni M.; Milanesi M.; Longoni P. D.;
demonstrated that the infusion of allogeneic MSCs into Matteucci P. et al. Human bone marrow stromal cells suppress T-
baboons was well tolerated in most animals and prolongs lymphocyte proliferation induced by cellular or nonspecific
mitogenic stimuli. Blood 99: 3838–3843; 2002.
the survival of allogeneic skin graft. Although the immu-
Dunnett S. B.; Bjorklund A; . Prospects for new restorative and
nomodulatory effects of MSCs have important implications neuroprotective treatments in Parkinson’s disease. Nature 399:
in transplantation biology, we do not know whether the A32–A39; 1999.
immunosuppressive effect of MSCs was affected by disease Fang B.; Liao L.; Shi M.; Yang S.; Zhao R. C. Multipotency of Flk1
+CD34+progenitors derived from human fetal bone marrow. J.
state. In this study, we demonstrated that PD MSCs are able
Lab. Clin. Med. 143: 230–240; 2005.
to inhibit the T cell proliferation stimulated in a mixed- Horwitz E. M.; Prockop D. J.; Fitzpatrick L. A. et al. Transplantability
lymphocyte culture or by incubation with mitogens. The and therapeutic effects of bone marrow-derived mesenchymal
immunosuppressive effect was similar between PD MSCs cells in children with osteogenesis imperfecta. Nat. Med. 5: 309–
313; 1999.
and normal adult MSCs, suggesting that PD MSCs still
Jankovic J. Levodopa strengths and weaknesses. Neurology 58: S19–
preserved the immunosuppressive effects. S32; 2002.
In summary, we described for the first time the Jiang Y.; Jahagirdar B. N.; Reinhardt R. L. et al. Pluripotency of
biological characteristics of MSCs derived from PD mesenchymal stem cell derived from adult marrow. Nature 418:
41–49; 2002.
patients’ bone marrow. Our results showed that PD-derived
Kasten P.; Vogel J.; Luginbühl R.; Niemeyer P.; Weiss S.; Schneider S.
MSCs are similar to normal MSCs in phenotype, morphol- et al. Influence of platelet-rich plasma on osteogenic differenti-
ogy, and multidifferentiation capacity. Moreover, PD- ation of mesenchymal stem cells and ectopic bone formation in
FUNCTION OF AUTOLOGOUS MESENCHYMAL STEM CELL 177

calcium phosphate ceramics. Cells Tissues Organs 183: 68–79; embryonic stem cells. Proc. Natl. Acad. Sci. USA 101: 12543–
2006. 12548; 2004.
Kawasaki H.; Mizuseki K.; Nishikawa S.; Kaneko S.; Kuwana Y.; Pittenger M. F.; Mackay A. M.; Beck S. C. et al. Multilineage
Nakanishi S. et al. Induction of midbrain dopaminergic neurons potential of adult human mesenchymal stem cells. Science 284:
from ES cells by stromal cell-derived inducing activity. Neuron 143–147; 1999.
28: 3140; 2000. Quinn N. P. Parkinson’s disease: clinical features. Baillieres Clin.
Koc O.; Lazarus H. Mesenchymal stem cells heading to the clinic. Neurol. 6: 1–13; 1997.
Bone Marrow Transplant. 27: 235–239; 2001. Reyes M.; Lund T.; Lenvik T. et al. Purification and ex vivo expansion
Kopen G. C.; Prockop D. J.; Phinney D. G. Marrow stromal cells of postnatal human marrow mesodermal progenitor cells. Blood
migrate throughout forebrain and cerebellum, and they differen- 98: 2615–2625; 2001.
tiate into astrocytes after injection into neonatal mouse brains. Sakurada K.; Ohshima-Sakurada M.; Palmer T. D.; Gage F. H. Nurr1,
Proc. Natl. Acad. Sci. USA 96: 10711–10716; 1999. an orphan nuclear receptor, is a transcriptional activator of
Krampera M.; Glennie S.; Dyson J.; Scott D.; Laylor R.; Simpson E. endogenous tyrosine hydroxylase in neural progenitor cells
et al. Bone marrow mesenchymal stem cells inhibit the response derived from the adult brain. Development 126: 4017–4026; 1999.
of naive and memory antigen-specific T cells to their cognate Sanchez-Ramos J.; Song S.; Cardozo-Pelaez F.; Hazzi C.; Stedeford
peptide. Blood 101: 3722–3729; 2003. T.; Willing A. et al. Adult bone marrow stromal cells differentiate
Le Blanc K.; Rasmusson I.; Sundberg B. et al. Treatment of severe into neural cells in vitro. Exp. Neurol. 164: 247–256; 2000.
acute graft-versus-host disease with third party haploidentical Seta Y.; Toyono T.; Takeda S.; Toyoshima K. Expression of Mash1 in
mesenchymal stem cells. Lancet 363: 1439–1441; 2004. basal cells of rat circumvallate taste buds is dependent upon
Lee S. H.; Lumelsky N.; Studer L.; Auerbach J. M.; McKay R. D. G. gustatory innervation. FEBS Lett. 444: 4346; 1999.
Efficient generation of midbrain and hindbrain neurons from Tse W. T.; Pendleton J. D.; Beyer W. M.; Egalka M. C.; Guinan E. C.
mouse embryonic stem cells. Nat. Biotechnol 18: 675–679; 2000. Suppression of allogeneic T-cell proliferation by human marrow
Makino S.; Fukuda K.; Miyoshi S. et al. Cardiomyocytes can be stromal cells: implications in transplantation. Transplantation 75:
generated from marrow stromal cells in vitro. J. Clin. Invest. 103: 389–397; 2003.
697–705; 1999. Woodbury D; Schwarz E. J.; Prockop D. J.; Black I. B. Adult rat and
Mezey E.; Chandross K. J.; Harta G.; Maki R. A.; Mckercher S. R. human bone marrow stromal cells differentiate into neurons. J.
Turning blood into brain; cells bearing neuronal antigens Neurosci. Res. 61: 364–370; 2000.
generated in vivo from bone marrow. Science 290: 1779–1782; Zhang H.; Huang Z.; Xu Y.; Zhang S. Differentiation and neurological
2000. benefit of the mesenchymal stem cells transplanted into the rat
Minguell J. J.; Conget P.; Erices A. Biology and clinical utilization of brain following intracerebral hemorrhage. Neurol. Res. 28: 104–
mesenchymal progenitor cells. Braz. J. Med. Biol. Res. 33: 881– 112; 2006.
887; 2000. Zhao L. R.; Duan W. M.; Reyes M. et al. Human bone marrow stem
Park S.; Lee K. S.; Lee Y. J.; Shin H. A.; Cho H. Y.; Wang K. C. et al. cells exhibit neural phenotypes and ameliorate neurological
Generation of dopaminergic neurons in vitro from human deficits after grafting into the ischemic brain of rats. Exp. Neurol.
embryonic stem cells treated with neurotrophic factors. Neurosci. 174: 11–20; 2002.
Lett. 359: 99–103; 2004. Zheng J. L.; Shou J.; Guillemot F.; Kageyama R.; Gao W. Q. Hes1 is a
Perrier A. L.; Tabar V.; Barberi T.; Rubio M. E.; Bruses J.; Topf N. negative regulator of inner ear hair cell differentiation. Development
et al. Derivation of midbrain dopamine neurons from human 127: 4551–4560; 2000.

You might also like