You are on page 1of 6

Int. J.

Devl Neuroscience 37 (2014) 94–99

Contents lists available at ScienceDirect

International Journal of Developmental Neuroscience


journal homepage: www.elsevier.com/locate/ijdevneu

Directing mouse embryonic neurosphere differentiation toward an


enriched neuronal population
Ema F. Torrado a,1 , Cátia Gomes a,1 , Gisela Santos a , Adelaide Fernandes a,b , Dora Brites a,b,∗ ,
Ana S. Falcão a,b,∗
a
Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisbon, Portugal
b
Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisbon,
Portugal

a r t i c l e i n f o a b s t r a c t

Article history: Neural stem cells (NSC) are self-renewing multipotent cells that have emerged as a powerful tool to
Received 22 April 2014 repair the injured brain. These cells can be cultured as neurospheres, which are floating aggregates of
Received in revised form 30 June 2014 neural stem/progenitor cells (NSPCs). Despite their high clonal expansion capacity, it has been suggested
Accepted 1 July 2014
that in neurospheres, only a small percentage of cells are capable of proliferation and that this system
is not efficient in terms of neurogenic competence. Thus, our aim was to develop a neurosphere culture
Keywords:
method with a highly proliferative stem/progenitor cell population and particularly with a prominent
Mouse neural stem cells
neurogenic potential, surpassing some of the claimed weaknesses of the neurosphere assay. In our model,
Neuronal differentiation
Neural progenitor cells
mouse neurospheres were harvested from neural tissue at E15 and after only 4 days in vitro (DIV), we
Mouse cortex E15 neurospheres have achieved highly proliferative primary neurospheres (81% Sox2 and 76% Ki67 positive cells) and a
rather low number of cells expressing glial and neuronal markers (∼10%). After inducing differentiation,
we have attained an enriched neuronal population (45% ␤-III-tubulin positive cells at 15 DIV). Using a
simple methodology, we have developed a NSPC model that can provide a valuable source of neuronal
precursors, thus offering a potential starting point for cell replacement therapies following CNS injury.
© 2014 ISDN. Published by Elsevier Ltd. All rights reserved.

1. Introduction However, these models have proven to not accurately recapitu-


late in vivo development processes of the nervous system, such as
In the last 20 years, neural stem cell (NSC) research has shown proliferation, migration, differentiation and synaptogenesis (Breier
to be a promising tool for studying and elucidating the mole- et al., 2010). NSC are multipotent cells that possess the ability to
cular and cellular properties of brain development, plasticity and self-renew and the potential to generate the three major cell types
regeneration, as well as an experimental basis for new biome- of the CNS, i.e., neurons, astrocytes and oligodendrocytes (Conti
dical applications. In fact, NSC can offer a renewable source for and Cattaneo, 2010). Since the first studies from Temple (1989),
cell replacement therapy in neurodegenerative diseases (Suter and NSC have been isolated from several brain regions and there are
Krause, 2008) and may be used in assays aimed to test promising many assays for their isolation and expansion (Conti and Cattaneo,
therapeutic compounds for pharmaceutical and neurotoxic prop- 2010). Among these, the isolation of NSC through the neurosphere
erties (Fritsche et al., 2011). In the past, the in vitro systems that formation assay is one of the most widely used for NSC prolif-
were commonly used to study neurodevelopmental processes were eration. Neurospheres were first described in 1992, using both
restricted to transformed cell lines from humans (e.g. SH-SY5Y) or adult and the embryonic CNS (Reynolds and Weiss, 1992; Reynolds
rodents (PC12), as well as primary cultures obtained from rodent et al., 1992), and are defined as three dimensional structures that
central nervous system (CNS) or fetal tissue derived from abortions. grow as floating aggregates and are composed by a heteroge-
neous mixture of multipotent NSC, proliferating neural progenitor
cells (NPC) and postmitotic neurons and glia (Jensen and Parmar,
2006). The neurosphere system has been used extensively since it
∗ Corresponding authors at: Research Institute for Medicines (iMed.ULisboa),
is thought to provide a three-dimensional environment that can
Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto,
mimic the neurogenic niche (Giachino et al., 2009). In fact, the neu-
1649-003 Lisbon, Portugal. Tel.: +351 2179464900; fax: +351 217946491.
E-mail addresses: dbrites@ff.ul.pt (D. Brites), asfalcao@ff.ul.pt (A.S. Falcão). rosphere system is considered as a valuable in vitro model to study
1
These authors contributed equally to this paper. neurodevelopmental processes, as it is maintained under defined

http://dx.doi.org/10.1016/j.ijdevneu.2014.07.001
0736-5748/© 2014 ISDN. Published by Elsevier Ltd. All rights reserved.
E.F. Torrado et al. / Int. J. Devl Neuroscience 37 (2014) 94–99 95

serum-free conditions where the environmental cues are limited Merck Millipore), a component of microtubules regarded as a neuron-specific
to those of the adjacent cells (Jensen and Parmar, 2006). Another marker; the chondroitin sulfate proteoglycan NG2 (1:200, AB5320, Merck Millipore),
that stains for oligodendrocyte precursor cells; and the myelin basic protein (MBP,
important advantage of the neurosphere system is that it has a great
1:200, MAB387, Merck Millipore), which is a marker for mature oligodendrocytes.
passage potential as compared to adherent monocultures, enabling We have also performed a proliferation assay, by using a specific antibody against
large scale in vitro expansion of neural stem and progenitor cells for the nuclear protein Ki67 (1:25, sc-7846, Santa Cruz Biotechnology, Dallas, USA). For
potential drug screening and cell replacement therapy (Sun et al., each coverslip, nuclei were stained with Hoechst dye 33258 (Sigma). All images
2011). However, this culture method also has some disadvantages. were acquired using a fluorescence microscope (AxioScope A1, Zeiss, Oberkochen,
Germany) with an integrated camera (AxioCamHRm, Zeiss). Immune-positive cells
Thus, although it can mirror the cell heterogeneity of the in vivo NSC of each type and total cell number (at least 1000 cells per slide) were counted and
niche, the three-dimensional structure of neurospheres shows no the values were presented as percentage of positive cells for each staining ±SEM.
spatial cellular organization in terms of cell types that are typical of
these niches. Another weakness of the neurosphere system is that 2.3. Differentiation of neurospheres
there are some studies claiming that only a small percentage of cells
After 4 DIV under proliferating conditions, another set of neurospheres was
within a neurosphere are capable of proliferation (Louis et al., 2008;
induced to differentiate by culturing dissociated neurospheres in Neurobasal
Moors et al., 2009; Pastrana et al., 2011). In addition, it is described medium (Invitrogen) supplemented with 0.5 mM l-glutamine, 2% B-27 Supplement
that this model is not particularly efficient in terms of neurogenic (Invitrogen), 30 mM glucose, 50 ng/mL bovine serum albumin, 1% fetal bovine serum
competence (Breier et al., 2010; Conti et al., 2005). and 1% AB/AM in tissue culture plates (with coverslips) pre-coated with PDL, at
1 × 105 cells/cm2 (adapted from Smith et al., 2003). Every 3 days, half of old medium
Thus, our aim was to develop a mouse neurosphere culture
was removed by aspiration and replaced by the same volume of fresh medium. Cells
method that could obviate some of the flaws above indicated, were fixed at 3, 9 and 15 DIV under differentiating conditions, and processed for
i.e., with a high proliferating stem/progenitor cell population and characterization by immunocytochemistry, using the same neural markers previ-
an efficient neurogenic potential after inducing differentiation. ously indicated for neurospheres.
Starting from E15 mouse cortices, we have obtained highly prolif-
erative neurospheres after 4 days in vitro (DIV) that also expressed 2.4. Statistical analysis

a low amount of glial and neuronal markers. After inducing neural Results of at least three different experiments, performed in duplicate, were
differentiation with the culture medium that is used in primary expressed as mean ± SEM. Data was statistically evaluated using the two-tailed t
neuronal cultures (Falcão et al., 2007), this system revealed to test on the basis of equal or unequal variance, as appropriate. We considered P < 0.05
contain a high percentage of neuronal cells relatively to that of to be statistically significant.
astrocytes and oligodendrocytes, as compared to some of the
currently available protocols. Thus, this simple methodological 3. Results and discussion
approach may be potentially useful when considering therapeu-
tic applications of neuronal replacement in neurodegenerative The RHB-A® medium used for neurospheres proliferation and
diseases and CNS injury. expansion allowed a fast formation of the floating aggregates as
soon as 2 DIV after plating. In fact, this medium was previously
2. Materials and methods used for a more efficient production of NPC from embryonic stem
cells in monolayer cultures (Abranches et al., 2009). In addition, the
2.1. Animal procedures
presence of the growth factors EGF and bFGF also promoted sus-
Animal care followed the recommendations of European Convention for the Pro- tained self-renewing divisions of NSC (Conti et al., 2005). After 4
tection of Vertebrate Animals Used for Experimental and Other Scientific Purposes DIV, neurospheres were characterized in order to validate the het-
(Council Directive 86/609/EEC) and National Law 1005/92 (rules for protection of erogeneous composition of these structures. Since there are some
experimental animals). All animal procedures were approved by the Institutional
studies claiming that only a small percentage of cells within the
Animal Care and Use Committee.
neurosphere are capable of proliferation (Louis et al., 2008; Moors
et al., 2009; Pastrana et al., 2011), we have first determined the pro-
2.2. Neurospheres cultures and characterization
portion of stem/progenitor cells of these tridimensional structures
Neurospheres were isolated from CD1 mouse fetuses at embryonic day 15. by determining the number of Sox2 positive cells. Sox2 plays a cru-
Briefly, pregnant mice were euthanized by asphyxiation with CO2 and the fetuses cial role in embryonic development and in the developing brain its
were rapidly decapitated. After removal of meninges and white matter, the neocor- expression is first detected uniformly in the neural plate, in which
tices were collected in Hanks’ balanced salt solution without Ca2+ and Mg2+ (HBSS,
Invitrogen, Carlsbad, USA) and transferred to trypsin–EDTA (5 g/L trypsin–2 g/L
most of the cells are multipotent, controlling the expression of se-
EDTA, Sigma, St. Louis, USA) plus 1 U/mL DNAse I (Sigma) solution, for 30 min at 37 ◦ C. veral developmentally important genes (Wegner and Stolt, 2005).
Following trypsinization, cells were washed three times with HBSS, resuspended in In mouse embryo brains, it is mainly expressed in the expanding
a final volume of RHB-A® stem cell culture medium (SCS-SF-NB-01, StemCells Inc., cell population of the ventricular zone, in which NPC are generated
Newark, USA) and mechanically dissociated using a Pasteur pipette. Approximately
to subsequently establish the functional neocortex (Bani-Yaghoub
1 × 106 cells/mL were plated on tissue culture uncoated plates in the presence of
10 ng/mL murine epidermal growth factor (EGF, PeproTech, London, UK), 10 ng/mL et al., 2006). Furthermore, it is described that this transcription fac-
murine basic fibroblast growth factor (bFGF, PeproTech) and 1% antibiotic antimy- tor is essential for the maintenance of NSC, since inhibition of Sox2
cotic solution (AB/AM, Sigma), and maintained at 37 ◦ C in a humidified atmosphere signaling promotes an early exit from the cell cycle, while consti-
of 5% CO2 . Under these proliferating conditions, the cells grow as free-floating neuro- tutive expression inhibits neuronal differentiation (Graham et al.,
spheres. Proliferating medium was changed after 2 DIV by mechanical dissociation
of neurospheres and replating cells on uncoated tissue culture plates at a density of
2003). Generally, the majority of studies use Nestin as a NSC marker.
1 × 106 cells/mL. At 4 DIV, a set of neurospheres were trypsinized to obtain a suspen- However, this protein is also detected in immature neurons. In
sion of dissociated cells. These cells were then plated in tissue culture plates (with fact, it was referred that when green fluorescent protein-labeled
coverslips) pre-coated with poly-d-lysine (PDL, Sigma), and fixed after 4 h with 4% Nestin positive cells are sorted and tested for neurosphere forma-
paraformaldehyde (w/v) in PBS. This way, instead of having cells in the 3D structure
tion, less than 1% of the cells were able to form neurospheres, thus
of neurospheres, we have isolated cells that can be characterized by immunocyto-
chemistry. Since neurospheres are a heterogeneous population of cells composed suggesting that this marker is not NSC exclusive (Mignone et al.,
by NSC, NPC and differentiated cells, we have used different specific antibodies 2004). Our results (Fig. 1) demonstrate that about 81% of the cells
against: the Sox2 (Sex determining region of Y chromosome-related high motility that constitute the neurospheres were Sox2-positive and the Ki67
group box2) protein (1:500, ab5603, Merck Millipore, Darmstadt, Germany), a self- staining confirmed that most of these cells were in a proliferat-
renewal marker of neural stem/progenitor cells (NSPC); the glial fibrillary acidic
protein (GFAP, 1:100, MAB360, Merck Millipore), an astrocytic marker; the neu-
ing state (76 ± 2.7%). Compared with the 31% of Sox2-positive cells
ronal protein HuC/D (1:750, A-21271, Molecular Probes, Carlsbad, USA), an early obtained by (Park et al., 2007) in embryonic mice primary neuro-
postmitotic neuronal precursor marker; the ␤-III-tubulin protein (1:500, MAB1637, spheres, our procedure present a major advantage in allowing a
96 E.F. Torrado et al. / Int. J. Devl Neuroscience 37 (2014) 94–99

Fig. 1. Expression of undifferentiated and early post-mitotic neuronal cell markers in neurospheres derived from E15 mice cortices and in resulting differentiating cells
at 3, 9 and 15 days in vitro (DIV). (A) Representative results of one experiment are shown. Nuclei were stained with Hoechst dye (blue) and cells labeled for Sox2 (neural
stem/progenitor cells in green, upper panel) and HuC/D (early post-mitotic neurons in red, lower panel). Merged images of Sox2 and HuC/D are shown between the Sox2
and HuC/D columns and arrow heads represent cells that have positive double staining. Scale bar represents 20 ␮m. (B) Graph bars represent, for each developmental stage,
the percentage of cells (mean ± SEM) positive for Sox2 and HuC/D relatively to the total number of nuclei (at least 1000 cells per slide), from at least three independent
experiments, performed in duplicate. **P < 0.01 vs. the respective neural marker expression in neurospheres. (For interpretation of the references to color in this figure
legend, the reader is referred to the web version of the article.)

very high yield in NSPC content, corroborating once more the ben- the maximum value of 42% (P < 0.01). After this time point, the
efits of our proliferation medium and validating this protocol as percentage of HuC/D positive cells started to decrease although
a good source of NSPC for therapeutic applications, such as cellu- neuronal maturation was still occurring, which was observed by
lar transplantation. After inducing cell differentiation, there was a the increase in the number of ␤-III-tubulin positive cells (Fig. 2).
marked decrease in Sox2 staining (P < 0.01) to 49% at 3 DIV, 19% at Despite several differentiation protocols using embryonic mice
9 DIV and 14% at 15 DIV, as expected. During the differentiation primary neurospheres, where most of the works did not succeed
process, since neuronal cells do not express this transcription fac- in obtaining more than 30% of ␤-III-tubulin positive cells (Conti
tor, this labeling corresponds to persisting NSPC and proliferating and Cattaneo, 2010; Ahlenius et al., 2009; Cavallaro et al., 2008;
astrocytes that are referred to maintain Sox2 expression through- Smith et al., 2003; Ostenfeld et al., 2002), we have achieved 31%
out differentiation until they reach a quiescent state (Bani-Yaghoub of these cells at 9 DIV (P < 0.01) further increasing to 45% at 15
et al., 2006). DIV (P < 0.01), meaning a continuous process of neuronal matu-
Neurospheres also expressed HuC/D, which is an early postmi- ration and a promising source of viable neurons (Fig. 2). Higher
totic neuronal indicator (Fujiyama et al., 2009) and thus, a marker values of ␤-III-tubulin positive cells have been recently achieved
of neuronal precursors (Ghai et al., 2008). For a clearer perception by using a shaking purification method based on the differential
of the nature of these HuC/D positive cells, we have performed a substrate attachment properties of neuronal and glial cells in a dif-
double staining with Sox2 (Fig. 1A) and observed that there was ferentiating neural stem cell progeny culture (Azari et al., 2014),
a percentage of these HuC/D positive cells that also express the or by applying more complex and costly methodologies, such as a
NSC marker (i.e. 27 ± 4.7%, 26 ± 1.1%, 34 ± 3.4% and 42 ± 6.9% from purification step or cell sorting technology (Azari et al., 2011, 2012).
total HuC/D cells in neurospheres, and in cells at 3, 9 and 15 DIV of We consider that the high neurogenic potential of our neurosphere
differentiation, respectively). This means that some of the HuC/D model relies on the differentiation medium composition. This cul-
positive cells that have just exited the cell cycle also express mark- ture medium is the same we use to obtain pure primary cultures
ers of proliferation, and thus, we can identify this population as of neurons (Falcão et al., 2007), and where the presence of Neu-
early neuronal progenitor cells. After inducing differentiation we robasal medium associated with B-27 supplement were shown to
have observed a rapid increase in neuronal precursors at 3 DIV favor neuronal maturation and survival (Svendsen et al., 1995). In
(31% of HuC/D positive cells, P < 0.01) until 9 DIV, where it reached addition, due to the short proliferation period of our neurospheres,
E.F. Torrado et al. / Int. J. Devl Neuroscience 37 (2014) 94–99 97

Fig. 2. Expression of neuronal and glial cell markers in neurospheres derived from E15 mice cortices and in resulting differentiating cells at 3, 9 and 15 days in vitro (DIV).
(A) Representative results of one experiment are shown. Nuclei were stained with Hoechst dye (blue) and cells labeled for ␤-III-tubulin (neurons in green, upper panel) and
glial fibrillary acidic protein (GFAP; astrocytes in red, lower panel). Scale bar represents 20 ␮m. (B) Graph bars represent, for each developmental stage, the percentage of
cells (mean ± SEM) positive for ␤-III-tubulin and GFAP relatively to the total number of nuclei (at least 1000 cells per slide), from at least three independent experiments,
performed in duplicate. *P < 0.05 and **P < 0.01 vs. the respective neural marker expression in neurospheres. (For interpretation of the references to color in this figure legend,
the reader is referred to the web version of the article.)

we have prevented a loss of their neurogenic potential by avoiding progenitors was not accompanied by the mature, MBP-positive,
long-term expansion (Wright et al., 2006). myelin-generating, oligodendrocytes. This is not surprising since it
Regarding astrocytes, most studies have demonstrated GFAP is well known that maturation of oligodendrocytes only takes place
expression on these floating cell aggregates from mouse origin, at early postnatal life (Baumann and Pham-Dinh, 2001). In fact, it is
in contrast with those from human origin which evidence weak described that even if the neurosphere cultures were obtained from
immunoreactivity to anti-GFAP antibodies (Sun et al., 2008). We the adult mouse hippocampus, which is considered a NSC niche
found that the percentage of GFAP positive cells in neurospheres (Jhaveri et al., 2010), there is still no staining for this marker. NG2
was very similar to the one obtained for neuronal precursors (∼10%, and GFAP positive cells in percentages like our own were previously
Fig. 2), suggesting that these proliferating cells may have the same documented in other neurosphere protocol (Fernandez et al., 2009).
neurogenic and astrogliogenic potential. After differentiation, this Although the number of cells expressing MBP almost duplicated
glial phenotype reached a peak at 9 DIV (22%, P < 0.01) without any from 3 to 9 DIV, we assume that our culture medium is not appro-
further increase, while neuronal differentiation is still occurring. priate to further increment or at least to maintain constant the
To this it may have accounted the fact that our culture medium number of MBP positive cells. The bovine serum albumin present in
was not supplemented with higher concentrations of fetal bovine our differentiating protocol, as well as the one in the B-27 supple-
serum. Indeed, increased levels of this supplement in the differen- ment, by containing some bone morphogenic protein-like activity
tiation medium (at least ten times higher) were indicated to favor (Clark and Ding, 2007), may have contributed to inhibit further
the differentiation of stem cells into astrocytes (astrogliogenesis) oligodendrocyte precursor cell maturation (See et al., 2004). Nev-
and consequently, an increase in GFAP positive cells (Brunet et al., ertheless, if desirable, both NG2 and MBP enriched cultures may be
2004; Abe et al., 2006). obtained by magnetic cell sorting (Cizkova et al., 2009).
Neurospheres have also the ability to differentiate into oligo- In conclusion, our data support that cell fate decisions of CNS
dendrocytes, the myelin-forming cells of the CNS. NG2 positive cells cells may be controlled efficiently by the presence of some extrin-
were shown to preferentially occupy the outer zone of the neu- sic factors in proliferating/differentiating conditions (Johe et al.,
rosphere (Mokry et al., 2008) and in our model they represented 1996). Based on this statement, we have adapted some existing
12% of all cells (Fig. 3). The presence of these oligodendrocytes protocols regarding neurospheres proliferation (Abranches et al.,
98 E.F. Torrado et al. / Int. J. Devl Neuroscience 37 (2014) 94–99

Fig. 3. Expression of progenitor and mature oligodendrocytes markers in neurospheres derived from E15 mice cortices and in resulting differentiating cells at 3, 9 and 15 days
in vitro (DIV). (A) Representative results of one experiment are shown. Nuclei were stained with Hoechst dye (blue) and cells labeled for NG2 (oligodendrocyte progenitor
cells, upper panel) and myelin basic protein (MBP; mature oligodendrocytes, lower panel). Scale bar represents 20 ␮m. (B) Graph bars represent, for each developmental
stage, the percentage of cells (mean ± SEM) positive for NG2 and MBP relatively to the total number of nuclei (at least 1000 cells per slide), from at least three independent
experiments, performed in duplicate. **P < 0.01 vs. the respective neural marker expression in neurospheres. (For interpretation of the references to color in this figure legend,
the reader is referred to the web version of the article.)

2009) and differentiation (Smith et al., 2003) and developed a very grants from Fundação para a Ciência e a Tecnologia (FCT), Lisbon,
simple experimental procedure with highly proliferative primary Portugal (to DB). ASF holds a post-doctoral research assistant posi-
neurospheres that in differentiating conditions ensure an enhanced tion (C2007-FFUL/UBMBE/02/2011) granted by FCT. The funding
neurogenic potential. Since it is described that the proliferation of organization had no role in study design, data collection and ana-
NSPC as floating aggregates is not particularly efficient in terms lysis, decision to publish, or preparation of the manuscript.
of neurogenic competence, the method here described seems use-
ful for cell differentiation into an enriched neuronal population, as
well as for studying mechanisms that underlie neurogenesis and/or References
gliogenesis. More importantly, it may also be adequate to human
embryonic tissue application, as human neurosphere cultures have Abe, T., Takahashi, S., Suzuki, N., 2006. Metabolic properties of astrocytes differen-
tiated from rat neurospheres. Brain Res. 1101, 5–11.
a greater capacity to divide in vitro and to generate neuronal cells
Abranches, E., Silva, M., Pradier, L., Schulz, H., Hummel, O., Henrique, D., Bekman,
(Ostenfeld et al., 2002). In fact, one of the major obstacles to the E., 2009. Neural differentiation of embryonic stem cells in vitro: a road map to
use of NSPC in neuronal replacement therapy is the limited abil- neurogenesis in the embryo. PLoS ONE 4, e6286.
Ahlenius, H., Visan, V., Kokaia, M., Lindvall, O., Kokaia, Z., 2009. Neural stem
ity of these cells to generate sufficient number of neurons either
and progenitor cells retain their potential for proliferation and differentiation
in vitro or after transplantation in animal models of neurodegener- into functional neurons despite lower number in aged brain. J. Neurosci. 29,
ative disease and CNS damage (Smith et al., 2003). Therefore, our 4408–4419.
model supports neurospheres as a suitable system for regenera- Azari, H., Osborne, G.W., Yasuda, T., Golmohammadi, M.G., Rahman, M., Deleyrolle,
L.P., Esfandiari, E., Adams, D.J., Scheffler, B., Steindler, D.A., Reynolds, B.A., 2011.
tive efforts following injury or disease, opening new avenues for Purification of immature neuronal cells from neural stem cell progeny. PLoS ONE
transplantation experimental approaches of functional neuronal 6, e20941.
cells. Azari, H., Sharififar, S., Fortin, J.M., Reynolds, B.A., 2012. The neuroblast assay: an
assay for the generation and enrichment of neuronal progenitor cells from dif-
ferentiating neural stem cell progeny using flow cytometry. J. Vis. Exp. 62, 3712.
Azari, H., Sharififar, S., Darioosh, R.P., Fortin, J.F., Rahman, M., Reynolds, B.A., 2014.
Acknowledgments Purifying immature neurons from differentiating neural stem cell progeny using
a simple shaking method. Stem Cell Res. Ther. 4, 178.
Bani-Yaghoub, M., Tremblay, R.G., Lei, J.X., Zhang, D., Zurakowski, B., Sandhu, J.K.,
This work was supported by the strategic project PEst- Smith, B., Ribecco-Lutkiewicz, M., Kennedy, J., Walker, P.R., Sikorska, M., 2006.
OE/SAU/UI4013/2011-2014 and the PTDC/SAU-NEU/64385/2006 Role of Sox2 in the development of the mouse neocortex. Dev. Biol. 295, 52–66.
E.F. Torrado et al. / Int. J. Devl Neuroscience 37 (2014) 94–99 99

Baumann, N., Pham-Dinh, D., 2001. Biology of oligodendrocyte and myelin in the Louis, S.A., Rietze, R.L., Deleyrolle, L., Wagey, R.E., Thomas, T.E., Eaves, A.C., Reynolds,
mammalian central nervous system. Physiol. Rev. 81, 871–927. B.A., 2008. Enumeration of neural stem and progenitor cells in the neural colony-
Breier, J.M., Gassmann, K., Kayser, R., Stegeman, H., De, G.D., Fritsche, E., Shafer, T.J., forming cell assay. Stem Cells 26, 988–996.
2010. Neural progenitor cells as models for high-throughput screens of devel- Mignone, J.L., Kukekov, V., Chiang, A.S., Steindler, D., Enikolopov, G., 2004. Neural
opmental neurotoxicity: state of the science. Neurotoxicol. Teratol. 32, 4–15. stem and progenitor cells in nestin-GFP transgenic mice. J. Comp. Neurol. 469,
Brunet, J.F., Grollimund, L., Chatton, J.Y., Lengacher, S., Magistretti, P.J., Villemure, 311–324.
J.G., Pellerin, L., 2004. Early acquisition of typical metabolic features upon dif- Mokry, J., Karbanova, J., Filip, S., Cizkova, D., Pazour, J., English, D., 2008. Phenotypic
ferentiation of mouse neural stem cells into astrocytes. Glia 46, 8–17. and morphological characterization of in vitro oligodendrogliogenesis. Stem
Cavallaro, M., Mariani, J., Lancini, C., Latorre, E., Caccia, R., Gullo, F., Valotta, M., DeBi- Cells Dev. 17, 333–341.
asi, S., Spinardi, L., Ronchi, A., Wanke, E., Brunelli, S., Favaro, R., Ottolenghi, S., Moors, M., Rockel, T.D., Abel, J., Cline, J.E., Gassmann, K., Schreiber, T., Schuwald, J.,
Nicolis, S.K., 2008. Impaired generation of mature neurons by neural stem cells Weinmann, N., Fritsche, E., 2009. Human neurospheres as three-dimensional
from hypomorphic Sox2 mutants. Development 135, 541–545. cellular systems for developmental neurotoxicity testing. Environ. Health Per-
Cizkova, D., Cizek, M., Nagyova, M., Slovinska, L., Novotna, I., Jergova, S., Radonak, spect. 117, 1131–1138.
J., Hlucilova, J., Vanicky, I., 2009. Enrichment of rat oligodendrocyte progenitor Ostenfeld, T., Joly, E., Tai, Y.T., Peters, A., Caldwell, M., Jauniaux, E., Svendsen, C.N.,
cells by magnetic cell sorting. J. Neurosci. Methods 184, 88–94. 2002. Regional specification of rodent and human neurospheres. Brain Res. Dev.
Clark, J.H., Ding, S., 2007. Chemically-defined culture of embryonic stem cells. In: Sul- Brain Res. 134, 43–55.
livan, C., Chad, A.C., Eggan, K. (Eds.), Human Embryonic Stem Cells: The Practical Park, J.H., Ahn, J.I., Kim, S.Y., Park, K.S., Lee, Y.D., Yamaguchi, M., Chung, H.J., 2007.
Handbook, vol. 918. Wiley, West Sussex, pp. 81–90. Genetic modification does not affect the stemness of neural stem cells in nestin
Conti, L., Cattaneo, E., 2010. Neural stem cell systems: physiological players or promoter-GFP transgenic mice. Neurosci. Lett. 421, 185–190.
in vitro entities? Nat. Rev. Neurosci. 11, 176–187. Pastrana, E., Vargas, V.S., Doetsch, F., 2011. Eyes wide open: a critical review of
Conti, L., Pollard, S.M., Gorba, T., Reitano, E., Toselli, M., Biella, G., Sun, Y., Sanzone, sphere-formation as an assay for stem cells. Cell Stem Cell 8, 486–498.
S., Ying, Q.L., Cattaneo, E., Smith, A., 2005. Niche-independent symmetrical self- Reynolds, B.A., Tetzlaff, W., Weiss, S., 1992. A multipotent EGF-responsive striatal
renewal of a mammalian tissue stem cell. PLoS Biol. 3, e283. embryonic progenitor cell produces neurons and astrocytes. J. Neurosci. 12,
Falcão, A.S., Silva, R.F.M., Pancadas, S., Fernandes, A., Brito, M.A., Brites, D., 2007. 4565–4574.
Apoptosis and impairment of neurite network by short exposure of immature Reynolds, B.A., Weiss, S., 1992. Generation of neurons and astrocytes from isolated
rat cortical neurons to unconjugated bilirubin increase with cell differentiation cells of the adult mammalian central nervous system. Science 255, 1707–1710.
and are additionally enhanced by an inflammatory stimulus. J. Neurosci. Res. 85, See, J., Zhang, X., Eraydin, N., Mun, S.B., Mamontov, P., Golden, J.A., Grinspan, J.B.,
1229–1239. 2004. Oligodendrocyte maturation is inhibited by bone morphogenetic protein.
Fernandez, M., Paradisi, M., Del, V.G., Giardino, L., Calza, L., 2009. Thyroid hormone Mol. Cell. Neurosci. 26, 481–492.
induces glial lineage of primary neurospheres derived from non-pathological Smith, R., Bagga, V., Fricker-Gates, R.A., 2003. Embryonic neural progenitor cells: the
and pathological rat brain: implications for remyelination-enhancing therapies. effects of species, region, and culture conditions on long-term proliferation and
Int. J. Dev. Neurosci. 27, 769–778. neuronal differentiation. J. Hematother. Stem Cell Res. 12, 713–725.
Fritsche, E., Gassmann, K., Schreiber, T., 2011. Neurospheres as a model for develop- Sun, T., Wang, X.J., Xie, S.S., Zhang, D.L., Wang, X.P., Li, B.Q., Ma, W., Xin, H., 2011.
mental neurotoxicity testing. Methods Mol. Biol. 758, 99–114. A comparison of proliferative capacity and passaging potential between neu-
Fujiyama, T., Yamada, M., Terao, M., Terashima, T., Hioki, H., Inoue, Y.U., Inoue, T., ral stem and progenitor cells in adherent and neurosphere cultures. Int. J. Dev.
Masuyama, N., Obata, K., Yanagawa, Y., Kawaguchi, Y., Nabeshima, Y., Hoshino, Neurosci. 29, 723–731.
M., 2009. Inhibitory and excitatory subtypes of cochlear nucleus neurons are Sun, Y., Pollard, S., Conti, L., Toselli, M., Biella, G., Parkin, G., Willatt, L., Falk, A.,
defined by distinct bHLH transcription factors, Ptf1a and Atoh1. Development Cattaneo, E., Smith, A., 2008. Long-term tripotent differentiation capacity of
136, 2049–2058. human neural stem (NS) cells in adherent culture. Mol. Cell. Neurosci. 38,
Ghai, K., Stanke, J.J., Fischer, A.J., 2008. Patterning of the circumferential marginal 245–258.
zone of progenitors in the chicken retina. Brain Res. 1192, 76–89. Suter, D.M., Krause, K.H., 2008. Neural commitment of embryonic stem cells:
Giachino, C., Basak, O., Taylor, V., 2009. Isolation and manipulation of mammalian molecules, pathways and potential for cell therapy. J. Pathol. 215, 355–368.
neural stem cells in vitro. Methods Mol. Biol. 482, 143–158. Svendsen, C.N., Fawcett, J.W., Bentlage, C., Dunnett, S.B., 1995. Increased survival of
Graham, V., Khudyakov, J., Ellis, P., Pevny, L., 2003. SOX2 functions to maintain neural rat EGF-generated CNS precursor cells using B27 supplemented medium. Exp.
progenitor identity. Neuron 39, 749–765. Brain Res. 102, 407–414.
Jensen, J.B., Parmar, M., 2006. Strengths and limitations of the neurosphere culture Temple, S., 1989. Division and differentiation of isolated CNS blast cells in microcul-
system. Mol. Neurobiol. 34, 153–161. ture. Nature 340, 471–473.
Jhaveri, D.J., Mackay, E.W., Hamlin, A.S., Marathe, S.V., Nandam, L.S., Vaidya, V.A., Wegner, M., Stolt, C.C., 2005. From stem cells to neurons and glia: a Soxist’s view of
Bartlett, P.F., 2010. Norepinephrine directly activates adult hippocampal pre- neural development. Trends Neurosci. 28, 583–588.
cursors via beta3-adrenergic receptors. J. Neurosci. 30, 2795–2806. Wright, L.S., Prowse, K.R., Wallace, K., Linskens, M.H., Svendsen, C.N., 2006. Human
Johe, K.K., Hazel, T.G., Muller, T., Dugich-Djordjevic, M.M., McKay, R.D., 1996. Single progenitor cells isolated from the developing cortex undergo decreased neuro-
factors direct the differentiation of stem cells from the fetal and adult central genesis and eventual senescence following expansion in vitro. Exp. Cell Res. 312,
nervous system. Genes Dev. 10, 3129–3140. 2107–2120.

You might also like