Advances in Bioscience and Biotechnology, 2013, 4, 853-859 ABB
http://dx.doi.org/10.4236/abb.2013.49113 Published Online September 2013 (http://www.scirp.org/journal/abb/)
Differentiation of neuronal cells using a murine embryonic
stem cell-based method
Uthayashanker R. Ezekiel1,2
1Department of Clinical Laboratory Science, Doisy College of Health Sciences, Saint Louis University Medical Center, Saint Louis,
USA
2GeneProTech, Inc., Saint Louis, USA
Email: uezekiel@slu.edu
Received 24 June 2013; revised 25 July 2013; accepted 10 August 2013
Copyright © 2013 Uthayashanker R. Ezekiel. This is an open access article distributed under the Creative Commons Attribution Li-
cense, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
The differentiation and screening methodology pro-
posed here is an efficient in vitro system to screen and
study effects of small molecules and bioagents and is
an alternative to studies that use live animals and
embryos. The method is based on engineering a stable
murine embryonic stem (ES) cell line expressing line-
age-specific promoters that drive selection and re-
porter genes. Additionally, uniform embryoid bodies
(EBs) are used for differentiation studies that allow
synchronous differentiation. The reporter and selec-
tion marker genes are expressed only in lineages
where the promoter is functional. The differentiated
cell type can be identified by reporter gene expression
and the selection marker can be used for selective
enrichment of that particular cell population. The
method described here is useful in screening small
molecules or bioagents that can differentiate stem
cells into particular lineages or cell types. Identified
compounds are useful in areas such as stem cell-based
regenerative medicine and therapeutics. The method
described here has been applied to neuronal cell dif-
ferentiation.
Keywords: Stem Cell; Embryoid Bodies; Differentiation
1. INTRODUCTION
Embryonic stem (ES) cells are pluripotent and derived
from an early embryonic stage of development. When
cultured in the presence of leukocyte inhibitory factor
(LIF), ES cells remain undifferentiated. Removal of LIF
and regulation of culture conditions lead to ES cell dif-
ferentiation into specific cell types [1]. Differentiation of
ES cells into different cell lineages is of great importance
in regenerative medicine. Differentiation into specific
cell type(s) and enrichment of the differentiated cell type(s)
are critical for therapeutic application purposes. We de-
scribe a method to screen small molecules/bioagents that
differentiate ES cells to a particular lineage and enrich
for a differentiated cell type.
An ES cell line containing a reporter and a selection
marker has been engineered with both reporter and selec-
tion markers under the control of a specific promoter in
the form of a Promoter-Selection-Reporter cassette. The
reporter and selection marker genes are expressed only in
lineages where the promoter is functional. The differen-
tiated cell type is identified by analysis of reporter gene
expression and the importance of the selection marker is
selective enrichment of a particular cell population. Sev-
eral strategies exist to differentiate ES cells into neuronal
cell types. Many differentiation strategies involve the
generation of embryoid bodies (EBs) that are aggregates
of ES cells in suspension [2-5]. Uniform-sized EBs have
been shown to have synchronous differentiation poten-
tials; therefore, it is critical to have uniform-size EBs for
screening purposes [6,7]. In the present study, neuron
differentiation has been used as a model system.
Many small molecules exert effects during differentia-
tion. Retinoic acid (RA) is an active metabolite of Vita-
min A, differentiates ES cells to neuronal lineages [8]
and acts through kinase-dependent pathways in neuro-
genesis[1,9]. Compounds that exert effects on signaling
pathways modulate stem cell differentiation. For exam-
ple, resveratrol, a natural compound present in grape skins,
acts on the extracellular signal-regulated kinase (ERK)-1
pathway in undifferentiated and differentiated neuron-
like cells [10]. Neuroprotective effects of Ginkgo biloba
extracts have been demonstrated in vitro and in vivo [11,
12]. An analog of Vitamin D having two double bonds in
the side chain causes differentiation of MG-63 human
osteosarcoma cells into osteoblasts [13]. Rho-associated
coiled-coil forming protein kinase (ROCK) inhibitor Y-
OPEN ACCESS
U. R. Ezekiel / Advances in Bioscience and Biotechnology 4 (2013) 853-859
854
27632 modulates stem cell differentiation [14]. Extracel-
lular signal-regulated kinase (ERK) 1/2 phosphorylation
inhibitor U0126 has been used to show ERK pathway
involvement in early neuronal differentiation of ES cells
[15]. In the present study, small molecules representing
natural compounds, kinase inhibitors or other factors pro-
moting neuronal differentiation have been used.
2. MATERIALS AND METHODS
2.1. Construction of Targeting Vector with
Neuron-Specific
Promoter-Reporter-Selection Cassette
An 844 bp DNA fragment upstream of the transcription
initiation start site for the necdin gene contains neuron-
restricted promoter activity [16]. For the present study,
this neuron-restrictive promoter region of the necdin
gene was cloned from a mouse genomic DNA by PCR
amplification of a 951 bp of DNA fragment upstream of
the translation initiation site [17,18]. The primers for
necdin amplification were chosen from the mouse necdin
sequence available from GenBank (accession number
D76440). The forward primer corresponded to nucleotide
numbers 1-26 (5’GATCATTTTC CACTAGAATC TTA
ACG3’) and the reverse primer corresponded to nu-
cleotide numbers 956-937 (5’TCTGATCCGA AGGCGC
AGAC3’) [16]: a PCR reaction was done using Pwo
DNA polymerase and the reaction was performed ac-
cording to the manufacturer’s guidelines (Roche Applied
Science). Plasmid pUC19 was modified with restriction
sites by sequential ligation of oligolinkers and a neomy-
cin phosphotransferase (Neo) gene under the control of
phosphoglycerate kinase promoter (PGK-1) was cloned.
The presence of the Neo gene allows selection of stably
transfected ES cells that confer resistance to antibiotic
G418 [19]. To the Neo cloned plasmid vector the fol-
lowing DNA fragments were cloned sequentially: ampli-
fied DNA fragment (956 bp, promoter); the antibiotic
resistance gene (919 bp, puromycin-N-acetyl-transferase
gene or Purr); internal ribosomal entry site (IRES); and
the reporter gene (β-galactosidase). The final plasmid
vector with Necdin-Promoter-Selection-Reporter cassette
was called pNec-SR vector (Figure 1).
2.2. ES Cell Maintenance and Stable ES Cell
Line Expressing Randomly Integrated
Necdin Promoter-Selection-Reporter
Cassette
ES cells derived from 129/SvJ mice were maintained in
culture on a layer of feeder cells consisting of mitomycin
C-treated primary mouse embryonic fibroblasts (MEF)
[20]. The ES cells were cultured in ES medium (Dul-
becco’s Modified Eagle Medium [DMEM]) supplemented
Figure 1. Targeting vector with Necdin promoter-selection-
reporter (pNec-SR). Plasmid pUC19 was modified with restric-
tion sites by sequential ligation of oligolinkers and the follow-
ing gene fragments were sequentially cloned: a neomycin gene
(Neo) under control of a phosphoglycerate kinase promoter
(PGK); the amplified necdin promoter DNA fragment; the anti-
biotic resistance gene, puromycin-N-acetyl-transferase gene (Purr)
IRES; and the reporter gene β-galactosidase.
with 15% serum (Hyclone, Logan, UT), 1000 units/ml
LIF (ESGRO from Chemicon, Temecula, CA), 1 mM
sodium pyruvate, 1 mM nonessential amino acids, 0.1
mM 2-mercaptoethanol, 25 units/ml penicillin and 25
μg/ml streptomycin. The ES cells were plated onto pre-
pared feeder layers with freshly prepared ES medium
and incubated in a humidified chamber (37˚C, 5% CO2).
Media were changed every other day for maintenance of
the ES cells.
A stable ES cell line with a randomly integrated necdin
Promoter-Selection-Reporter cassette was made. Briefly,
10 µg of pNec-SR vector was digested with 20 units of
SfiI at 50˚C for 2 hrs (New England Biolabs). After di-
gestion, the DNA was electrophoresed on agarose gel
and purified using QIAEX II gel extraction kit (Qiagen).
The linearized pNec-SR was electroporated (180 V, 500
µF) into ES cells and electroporated cells were plated
onto tissue culture dishes. ES cells were cultured (37˚C,
5% CO2) on an inactivated murine embryonic fibroblast
(MEF) feeder layer in standard ES cell culture media
containing LIF. After 24 h, G418 selection was per-
formed by addition of ES cell media containing G418
(400 μg/ml) and cells were allowed to grow for 5 days.
Only cells transfected with plasmid survived in the pres-
ence of G418. The surviving colonies were picked, ex-
panded and analyzed for the presence of β-galactosidase
and puromycin genes by PCR analysis. Six colonies were
positive: a single clone (ES-6) was selected as represent-
tative for further studies. As a predifferentiation step,
feeder cells were removed from the ES cell culture. To
accomplish feeder cell removal, ES cells were passaged
three times onto 0.1% gelatin-coated tissue culture plates
using the following protocol. The cells were trypsinized
(5 min) followed by trypsin inactivation (addition of an
equal volume of media and incubation in a humidified
chamber [15 min, 37˚C, 5% CO2]). After incubation,
most feeder cells remained attached to the plates whereas
Copyright © 2013 SciRes. OPEN ACCESS
U. R. Ezekiel / Advances in Bioscience and Biotechnology 4 (2013) 853-859 855
ES cells were in suspension. The ES suspension cells
were transferred to 0.1% gelatin-coated tissue culture
plates that supported ES cell growth and the presence/
absence of transferred feeder cells was microscopically
checked. This process was repeated a second and a third
time. After the third passage, all ES cells were com-
pletely devoid of feeder cells and were used for screen-
ing purposes.
2.3. EB Formation and Test Compound Addition
EBs were formed as described previously [7]. Single uni-
form sized EBs were formed in a multiwell plate, and
each well contained a single embryoid body (EB) [7]. An
ES-6 cell line containing a stably integrated necdin-
puromycin-IRES-β-galactosidase cassette was allowed to
form EB. Briefly, 1000 cells in 200 μl of media without
LIF were plated into 96-well PCR plates. On the third
day, the EBs were transferred to laminin-coated 96-well
tissue culture plates. The medium was changed on the
fourth day and appropriate small molecule compounds
were added with each compound being assessed in trip-
licate. Two positive controls, trans-retinoic acid (RA, at 1
µM, Sigma-Aldrich), nerve growth factor (NGF, at10
ng/ml, Sigma-Aldrich), no treatment control, vehicle con-
trol (dimethylsufoxide: DMSO, Sigma Aldrich), and five
small molecule compounds at 10 µM concentration (piper-
ine, curcumin, rosmarinic acid, U0126, LY294002; Sigma-
Aldrich) were used as screening compounds for neuron
differentiation. The cells were incubated for 48 h with
compounds. After 48 h, medium was removed, attached
cells were rinsed with phosphate buffered saline (PBS)
and medium was added. Cells were allowed to grow and
differentiate in media for 4 days and lysed on the fifth
day. Lysates were analyzed for β-galactosidase expres-
sion.
2.4. Expression Analysis of Beta-Galactosidase
and Neuronal Markers
The level of neuron differentiation was measured using a
sensitive chemiluminescent β-galactosidase assay (Clon-
tech). Since activity of the necdin promoter conferred
β-galactosidase expression, a quantitative measurement
of β-galactosidase activity [21] was performed using cell
lysate (cells exposed to compounds after day 5 of differ-
entiation). Activity was normalized to total protein con-
tent of the cell lysates.
The neuronal expression of beta-galactosidase and
neuronal markers was demonstrated by immunostaining.
For in situ immunofluorescence, cells were grown on
laminin-coated coverslips, rinsed with Tris-buffered sa-
line and fixed with 4% paraformaldehyde (in TBS, 10
min), washed with TBS and incubated with primary an-
tibody (in 1% BSA, 05% Triton X100: overnight, 4˚C).
After primary antibody incubation, the cells were washed
with TBS and incubated with fluorophore-conjugated sec-
ondary antibody in TBS containing Triton X100 (0.05%)
and 1% BSA (30 min). Cell types were visualized using
fluorescence microscopy.
After incubation with screening compounds, the EBs
were dissociated by gently pipetting and plated on laminin-
coated coverslips. On day 12, the cells were fixed and
double-immunostained with low-affinity nerve growth
factor receptor (LNGFR) also called p75 (p75, murine
monoclonal antibody; Sigma-Aldrich) and beta-galacto-
sidase (rabbit polyconal antibody; abcam) followed by
flurophore-conjugated secondary antibody (Alexa-568
conjugated to anti-mouse, Alexa-488 to goat anti-rabbit
respectively; Molecular Probes). Cells were stained with
4’,6-diamidino-2-phenylindole (DAPI; Molecular Probes)
to display cell nuclei.
2.5. Enrichment for Neurons Using Selection
Retinoic acid-differentiated cells were used to demon-
strate that the selection method enriched for neurons. EBs
were exposed to RA for 48 h and then the compound was
removed. Cells were allowed to grow on laminin-coated
coverslips for immunostaining or in 96 well microplate
for measurement of beta-galctosidase expression. After
retinoic acid exposure, cells were divided into two popu-
lations and changed to cell culture media. Two days after
growing in regular media, cells were subjected to puro-
mycin selection. One group was exposed to puromycin
(+) and another group, the no selection group, had no
puromycin (). After 12 d, cells were lysed and beta-
galactosidase activity was measured. Similar experiments
were carried out on laminin-coated coverslips for immu-
nocytochemistry. Cells were stained for the neuron marker,
microtubule associated protein 2 (MAP2), and the astro-
cyte marker, glial fibrilary acidic protein (GFAP). Fluo-
rescence stained cells were counted microscopically in at
least three fields.
3. RESULTS
3.1. Neuron-Restricted DNA Fragment of Necdin
Promoter Expresses Beta-Galctosidase
Activity in Neurons
Stably transfected ES cells containing the Necdin Pro-
moter-Selection-Reporter vector (pNec-SR vector, Fig-
ure 1) were differentiated into neuronal cells by retinoic
acid addition. To verify that necdin promoter drove the
beta-galactosidase reporter in neuronal cells, the retinoic
acid-differentiated cells grown on laminin-coated cover-
slips on day 12 were fixed and immunostained for the
neuronal marker, low-affinity nerve growth receptor
(LNGFR) and for beta-galactosidase (Figure 2(A)). As
expected, cells expressing LNGFR also expressed beta-
Copyright © 2013 SciRes. OPEN ACCESS
U. R. Ezekiel / Advances in Bioscience and Biotechnology 4 (2013) 853-859
856
galactosidase (Figure 2(A)) indicating that the necdin pro-
moter expressed beta-galactosidase in neurons. The same
area of Figure 2(A) is shown in bright field microscopy
in Figure 2(B), and the nuclei, visualized by DAPI stain,
of the cells are shown in Figure 2(C).
3.2. β-Galactosidase Activity as an Indicator of
Post-Mitotically Differentiated Neurons
The level of neuron differentiation was measured using a
sensitive chemiluminescent β-galactosidase assay (Clon-
tech). Since activity of the necdin promoter conferred
β-galactosidase expression, a quantitative measurement
of β-galactosidase activity was performed using cell lys-
ate. Compounds were removed after 48 h and cells were
allowed to differentiate for 4 d in laminin-coated 96 well
microplates. Activity was normalized to the total protein
content of the cell lysates. Two positive controls, NGF
and RA, and five small molecule compounds (piperine,
curcumin, rosmarinic acid, U0126, LY294002) were used
for screening (Figure 3). Fold increase was calculated
based on vehicle control (β-galactosidase activity). NGF
showed more neurogenesis (30-fold) than did RA (11-
fold), a result that was not surprising since NGF is a
stronger neurogenic growth factor than RA. Piperine had
a 3.3-fold increase in neurogenesis that was significantly
different from the positive controls. In this select set of 5
test compounds, the positive controls (RA and NGF) dem-
onstrated that the differentiated neurons are measurable
by beta-galactosidase reporter expression. However, since
none of the 5 test compounds displayed results equal to
or greater than the controls (NGF or RA) and since this
had been determined to be the screening strategy, the
interpretation of this EB-based screen was that none of
these test compounds had neurogenesis potential (Figure
3).
3.3. Selection Marker Enriches for Neurons
Exposure of differentiated cells to puromycin should have
allowed the survival of neurons because only neurons
expressed puromycin under control of the necdin pro-
moter. Cells were lysed on the 12th day with or without
Figure 2. The necdin promoter expresses beta-galactosidase in
neurons only (day 12 after selection). (A) Double immunostain-
ing using primary Ab for beta-galactosidase (murine mAb) and
low affinity NGF (p75) receptor (rabbit pAb) followed by
fluorophore-conjugated secondary Ab (Alexa-568 conjugated
goat anti-mouse; Alexa-488 goat anti-rabbit, respectively); (B)
Same area as A viewed with bright field microscopy; (C) Same
area as A stained with DAPI to display cell nuclei.
Figure 3. Effects of compounds on cell differentiation. Embry-
oid bodies were screened with controls and small molecule
compounds (n = 5 for NGF: n = 7 for RA with piperine and n =
3 for others). Fold increase was calculated based upon vehicle
alone treatment of control. Small molecule results indicated
significant (p < 0.05) differences of all agents compared with
NGF and RA. NGF = nerve growth factor. RA = retinoic acid.
PIP = piperine. CUR = curcumin. U01 = U0126. ROS = rosma-
rinic acid. LY = LY294002. Results are mean ± SEM. *p < 0.05.
puromycin treatment and the beta-galactosidase activity
was measured. As shown in Figure 4(A), a puromycin-
treated population exhibited greater beta-galactosidase
activity than an untreated population. Next, cells were
immunostained and the cells displaying marker expres-
sion (MAP2 for neurons and GFAP for astrocytes) were
counted. Percent positive cells based on neuron and as-
trocyte markers, MAP2 and GFAP, were evaluated in the
puromycin-treated verus untreated differentiated neuronal
cells. As shown in Figure 4(B), puromycin selection eli-
minated astrocytes but enriched for neurons.
4. DISCUSSION
The ability to differentiate ES cells in vitro allows for
their use as model systems for the study of developmen-
tal potential and also as valuable reagents for stem cell
therapeutic approaches [22-24]. The method proposed
here is an efficient in vitro system to screen and study
effects of small molecules and bioagents. A positive hit
criterion that identifies a compound as one exhibiting
differentiation potential is its having similar or higher
reporter expression level than that of positive controls. In
this present study, positive controls were retinoic acid
and nerve growth factor. None of the five compounds
used for neuronal differentiation produced reporter ex-
pression similar to or greater than the positive controls. If
compounds are identified that have equal or greater re-
porter expression than positive controls in a larger screen-
ing strategy, then these compounds can be further evalu-
ated for differentiation potential. Using appropriate cul-
Copyright © 2013 SciRes. OPEN ACCESS
U. R. Ezekiel / Advances in Bioscience and Biotechnology 4 (2013) 853-859
Copyright © 2013 SciRes.
857
Figure 4. Puromycin selection (day 12). (A) Beta-galactosidase activity measured in cells treated without ( selection) and with (+
selection) puromycin. A significant difference (p < 0.05) between selection (+) and no selection ( selection) was noted in cells with
beta-galactosidase activity (results are mean ± SEM. n = 3); (B) Cells were stained with markers for neurons (MAP2 a + b) and as-
trocytes (GFAP) and then number of cells was counted. At least 3 areas on each slide were counted. A significant (p < 0.05) differ-
ence was observed between selected and unselected neurons and between selected and unselected astrocytes. Results reported as
mean ± SEM. n = 3. *p < 0.05.
genesis process proceeds in a manner similar to how it
actually occurs in a developing embryo. As an example,
the drug thalidomide has anti-angiogenic effects in vivo;
in other words, it inhibits endothelial cell formation. This
anti-angiogenic effect was not observed in either an en-
dothelial cell culture system or a chorioallantoic mem-
brane assay; however, anti-angiogenesis was noted in an
EB-based assay [33]. Finding the anti-angiogenic effect
of thalidomide only in an EB culture system and not by
other screening methods strongly indicates the need to
recapitulate intact organism development in small mole-
cule screens.
ture conditions, ES cells differentiate and form EBs that
contain cells of hematopoietic, endothelial, muscular and
neuronal lineages [25-29]. Many aspects of lineage-spe-
cific differentiation programs observed within EBs re-
flect those found in the embryo indicating this model
system provides access to early cell populations that de-
velop in a normal fashion [2,30]. ES cells are also able to
spontaneously differentiate and generate various lineages
under appropriate conditions in cell culture [31,32]. Us-
ing this in vitro system, it is possible to screen many
compounds efficiently and, once a candidate molecule is
identified, it can be studied in detail to define mecha-
nisms by which it exerts its effect. For screening pur-
poses, uniform-sized EBs have been shown to have syn-
chronous differentiation potentials [6]. The reason for
generating uniform-sized EBs is that factors produced by
cells present in EBs may influence the differentiation
pathway; therefore, it is critical to have uniform-size EBs
for screening purposes [23,28].
The method described in this paper allows identifica-
tion of differentiated cells by reporter gene expression
and selective enrichment of that particular cell popula-
tion. The differentiated cell type is then identified by
analysis of reporter gene expression. The importance of
the selection marker is selective enrichment of that par-
ticular cell population. By creation of a panel of lineage-
specific promoter-reporter ES cell lines, screening of EBs
from these cell lines will provide an excellent system for
testing differentiation effects of small molecules into many
different cell lineages.
Effects of small molecules may be studied using neu-
rons formed by an EB differentiation method or by direct
differentiation from ES cells with no EB intermediate
step. Comparison studies of developmental patterns of
murine EBs versus in vivo murine embryos have been
performed using gene expression and tagged reporter
constructs [2]. Results indicate that prior to day 3, EBs in
suspension culture are equivalent developmentally to pre-
gastrulation-stage embryos (4.5 - 6.5 days post coitum
[d.p.c]). Between days 3 and 5, EBs contain cell types
present in embryos during gastrulation (6.5 - 7.5 d.p.c.).
After day 6, EBs are equivalent to embryos in the stage
of early organogenesis (7.5 d.p.c) [2]. The EB differentia-
tion method is superior, however, because the EB neuro-
5. ACKNOWLEDGEMENTS
The author sincerely thank the critical evaluation of the manuscript by
Rita M. Heuertz and Sally Triocmi. This study is supported by a grant
from NIH/NINDS (1R43NS046133).
REFERENCES
[1] Xian, H.Q. and Gottlieb, D.I. (2001) Peering into early
neurogenesis with embryonic stem cells. Trends in Neu-
OPEN ACCESS
U. R. Ezekiel / Advances in Bioscience and Biotechnology 4 (2013) 853-859
858
rosciences, 24, 685-686.
doi:10.1016/S0166-2236(00)01966-4
[2] Leahy, A., Xiong, J.W., Kuhnert, F. and Stuhlmann, H.
(1999) Use of developmental marker genes to define
temporal and spatial patterns of differentiation during
embryoid body formation. Journal of Experimental Zo-
ology, 284, 67-81.
doi:10.1002/(SICI)1097-010X(19990615)284:1<67::AID
-JEZ10>3.0.CO;2-O
[3] Bain, G. and Gottlieb, D.I. (1998) Neural cells derived by
in vitro differentiation of P19 and embryonic stem cells.
Perspectives on Developmental Neurobiology, 5, 175-
178.
[4] Okabe, S., Forsberg-Nilsson, K., Spiro, A.C., Segal, M.
and McKay, R.D. (1996) Development of neuronal pre-
cursor cells and functional postmitotic neurons from em-
bryonic stem cells in vitro. Mechanisms of Development,
59, 89-102. doi:10.1016/0925-4773(96)00572-2
[5] Wichterle, H. and Peljto, M. (2008) Differentiation of
mouse embryonic stem cells to spinal motor neurons.
Current Protocols in Stem Cell Biology, Chapter 1:
Unit-1H.
[6] Ng, E.S., Davis, R.P., Azzola, L., Stanley, E.G. and Ele-
fanty, A.G. (2005) Forced aggregation of defined num-
bers of human embryonic stem cells into embryoid bodies
fosters robust, reproducible hematopoietic differentiation.
Blood, 106, 1601-1603. doi:10.1182/blood-2005-03-0987
[7] Ezekiel, U.R., Muthuchamy, M., Ryerse, J.S. and Heuertz,
R. (2006) Single embryoid body formation in a multi-
well plate. Electronic Journal of Biotechnology, 10, 328-
335.
[8] Bain, G., Ray, W.J., Yao, M. and Gottlieb, D.I. (1996)
Retinoic acid promotes neural and represses mesodermal
gene expression in mouse embryonic stem cells in culture.
Biochemical and Biophysical Research Communications,
223, 691-694. doi:10.1006/bbrc.1996.0957
[9] Chaerkady, R., Kerr, C.L., Marimuthu, A., Kelkar, D.S.,
Kashyap, M.K., Gucek, M., Gearhart, J.D. and Pandey, A.
(2009) Temporal analysis of neural differentiation using
quantitative proteomics. Journal of Proteome Research, 8,
1315-1326. doi:10.1021/pr8006667
[10] Miloso, M., Bertelli, A.A., Nicolini, G. and Tredici, G.
(1999) Resveratrol-induced activation of the mitogen-ac-
tivated protein kinases, ERK1 and ERK2, in human neu-
ronblastoma SH-SY5Y cells. Neuroscience Letters, 264,
141-144. doi:10.1016/S0304-3940(99)00194-9
[11] Ahlemeyer, B. and Krieglstein, J. (2003) Neuroprotective
effects of Ginkgo biloba extract. Cellular and Molecular
Life Sciences, 60, 1779-1792.
doi:10.1007/s00018-003-3080-1
[12] Yoo, D.Y., Nam, Y., Kim, W., Yoo, K.Y., Park, J., Lee,
C.H., Choi, J.H., Yoon, Y.S., Kim, D.W., Won, M.H., et
al. (2011) Effects of Ginkgo biloba extract on promotion
of neurogenesis in the hippocampal dentate gyrus in
C57BL/6 mice. Journal of Veterinary Medical Science,
73, 71-76. doi:10.1292/jvms.10-0294
[13] Mahonen, A., Jaaskelainen, T. and Maenpaa, P.H. (1996)
A novel vitamin D analog with two double bonds in its
side chain. A potent inducer of osteoblastic cell differen-
tiation. Biochemical Pharmacology, 51, 887-892.
doi:10.1016/0006-2952(95)02242-2
[14] Watanabe, K., Ueno, M., Kamiya, D., Nishiyama, A.,
Matsumura, M., Wataya, T., Takahashi, J.B., Nishikawa,
S., Nishikawa, S., Muguruma, K., et al. (2007) A ROCK
inhibitor permits survival of dissociated human embry-
onic stem cells. Nature Biotechnology, 25, 681-686.
doi:10.1038/nbt1310
[15] Li, Z., Theus, M.H. and Wei, L. (2006) Role of ERK 1/2
signaling in neuronal differentiation of cultured embry-
onic stem cells. Development, Growth & Differentiation,
48, 513-523. doi:10.1111/j.1440-169X.2006.00889.x
[16] Uetsuki, T., Takagi, K., Sugiura, H., and Yoshikawa, K.
(1996) Structure and expression of the mouse necdin
gene. Identification of a postmitotic neuron-restrictive
core promoter. Journal of Biological Chemistry, 271,
918-924. doi:10.1074/jbc.271.2.918
[17] Kuo, C.H., Uetsuki, T., Kim, C.H., Tanaka, H., Li, B.S.,
Taira, E., Higuchi, H., Okamoto, H., Yoshikawa, K. and
Miki, N. (1995) Determination of a necdin cis-acting ele-
ment required for neuron specific expression by using
zebra fish. Biochemical and Biophysical Research Com-
munications, 211, 438-446. doi:10.1006/bbrc.1995.1833
[18] Maruyama, E. (1996) Biochemical characterization of
mouse brain necdin. Biochemical Journal, 314, 895-901.
[19] Tucker, K.L., Wang, Y., Dausman, J. and Jaenisch, R.
(1997) A transgenic mouse strain expressing four drug-
selectable marker genes. Nucleic Acids Research, 25,
3745-3746. doi:10.1093/nar/25.18.3745
[20] Brook, F.A. and Gardner, R.L. (1997) The origin and
efficient derivation of embryonic stem cells in the mouse.
Proceedings of the National Academy of Sciences of the
United States of America, 94, 5709-5712.
doi:10.1073/pnas.94.11.5709
[21] Martin, C.S., Wight, P.A., Dobretsova, A. and Bronstein,
I. (1996) Dual luminescence-based reporter gene assay
for luciferase and beta-galactosidase. Biotechniques, 21,
520-524.
[22] Xiong, J.W., Battaglino, R., Leahy, A. and Stuhlmann, H.
(1998) Large-scale screening for developmental genes in
embryonic stem cells and embryoid bodies using retrovi-
ral entrapment vectors. Developmental Dynamics, 212,
181-197.
doi:10.1002/(SICI)1097-0177(199806)212:2<181::AID-
AJA4>3.0.CO;2-D
[23] Laposa, R. (2011) Stem cells for drug screening. Journal
of Cardiovascular Pharmacology, 58, 240-245.
doi:10.1097/FJC.0b013e31821823f5
[24] Barbaric, I., Jones, M., Harley, D.J., Gokhale, P.J. and
Andrews, P.W. (2011) High-content screening for chemi-
cal modulators of embryonal carcinoma cell differentia-
tion and survival. Journal of Biomolecular Screening, 16,
603-617. doi:10.1177/1087057111406547
[25] Schmitt, R.M., Bruyns, E. and Snodgrass, H.R. (1991)
Hematopoietic development of embryonic stem cells in
vitro: Cytokine and receptor gene expression. Genes &
Development, 5, 728-740. doi:10.1101/gad.5.5.728
[26] Risau, W., Sariola, H., Zerwes, H.G., Sasse, J., Ekblom,
P., Kemler, R. and Doetschman, T. (1988) Vasculogene-
Copyright © 2013 SciRes. OPEN ACCESS
U. R. Ezekiel / Advances in Bioscience and Biotechnology 4 (2013) 853-859
Copyright © 2013 SciRes.
859
OPEN ACCESS
sis and angiogenesis in embryonic-stem-cell-derived em-
bryoid bodies. Development, 102, 471-478.
[27] Wang, R., Clark, R. and Bautch, V.L. (1992) Embryonic
stem cell-derived cystic embryoid bodies form vascular
channels: An in vitro model of blood vessel development.
Development, 114, 303-316.
[28] O’Shea, K.S. (1999) Embryonic stem cell models of de-
velopment. Anatomical Record, 257, 32-41.
doi:10.1002/(SICI)1097-0185(19990215)257:1<32::AID-
AR6>3.0.CO;2-2
[29] Kane, N.M., Xiao, Q., Baker, A.H., Luo, Z., Xu, Q. and
Emanueli, C. (2011) Pluripotent stem cell differentiation
into vascular cells: A novel technology with promises for
vascular re(generation). Pharmacology & Therapeutics,
129, 29-49. doi:10.1016/j.pharmthera.2010.10.004
[30] Vallier, L., Mancip, J., Markossian, S., Lukaszewicz, A.,
Dehay, C., Metzger, D., Chambon, P., Samarut, J. and
Savatier, P. (2001) An efficient system for conditional
gene expression in embryonic stem cells and in their in
vitro and in vivo differentiated derivatives. Proceedings
of the National Academy of Sciences of the United States
of America, 98, 2467-2472. doi:10.1073/pnas.041617198
[31] Doss, M.X., Koehler, C.I., Gissel, C., Hescheler, J. and
Sachinidis, A. (2004) Embryonic stem cells: A promising
tool for cell replacement therapy. Journal of Cellular and
Molecular Medicine, 8, 465-473.
doi:10.1111/j.1582-4934.2004.tb00471.x
[32] Odorico, J.S., Kaufman, D.S. and Thomson, J.A. (2001)
Multilineage differentiation from human embryonic stem
cell lines. Stem Cells, 19, 193-204.
doi:10.1634/stemcells.19-3-193
[33] Sauer, H., Gunther, J., Hescheler, J., and Wartenberg, M.
(2000) Thalidomide inhibits angiogenesis in embryoid
bodies by the generation of hydroxyl radicals. American
Journal of Pathology, 156, 151-158.
doi:10.1016/S0002-9440(10)64714-1