ArticlePDF Available

Neural stem cells lose telomerase activity upon differentiating into astrocytes

Authors:

Abstract and Figures

Serum-free mouse embryo (SFME) cells were established by D. Barnes et al., and are known to be a neural stem cell line, which differentiate into astrocytes upon treatment with TGF-beta. Therefore, SFME cells is thought to be a model well suited to analyze the differentiation mechanism of neural stem cells. Until now, we have investigated the regulation mechanisms of telomerase activity and telomere length in human cancer and normal cells. Telomerase is the enzyme responsible for the synthesis and maintenance of telomere repeats located at chromosomal ends and is normally expressed in embryonic and germline cells, but not in most normal cells. Here, using SFME cells, we attempted to analyze the regulation mechanism of telomerase activity in neural stem cells and to detect a change upon differentiation into astrocytes. When SFME cells were cultured in the presence of TGF-beta, cells showed anelongated morphology and decreased its growth to 50% of control culture. Cells also expressed the glial fibrillary acidic protein (GFAP), a marker for astrocytes,indicating that TGF-beta induced differentiation in SFME cells from neural stem cells into astrocytes. At the same time,TGF-beta also inhibited telomerase activity and repressed the expression of the mouse telomerase reverse transcriptase(mTERT), demonstrating that SFME cells was vested with a finite replicative life span upon treatment with TGF-beta. To understand the mechanisms regulating mTERT levels during differentiation into astrocytes, we have estimated the expression level of c-myc, which is known to be a key molecule in activating the TERT promoter. As a result, TGF-beta-treated SFME cells were shown to repress the expression of c-myc. Furthermore, promoter analysis, using the 5'-region of the mTERT gene, which possess two E-box elements bound to c-Myc/Max, demonstrated that mTERT promoter activity greatly decreased in TGF-beta-treated SFME cells as compared to non-treated SFME cells. These suggest that c-myc might play a critical role in the expression of mTERT, and that down-regulation of c-myc dependent upon the astrocytic differentiation in SFME cells might cause the repression of mTERT in TGF-beta-treated SFME cells.
Content may be subject to copyright.
Cytotechnology 36: 137–144, 2001.
© 2001 Kluwer Academic Publishers. Printed in the Netherlands. 137
Neural stem cells lose telomerase activity upon differentiating into
astrocytes
Takumi Miura1, Yoshinori Katakura1, Katsuhiko Yamamoto1, Norihisa Uehara1, Toshie
Tsuchiya2, Eun-Ho Kim3& Sanetaka Shirahata1
1Department of Genetic Resources Technology, Kyushu University, Fukuoka 812-8581, Japan; 2Division of
Medical Devices, National Institution of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan;
3Graduate School of Biotechnology, Korea University, Seoul 136-701, Korea
(Author for correspondence; E-mail: katakura@grt.kyushu-u.ac.jp; Fax: +81 92 642 3050)
Received 12 December 2000; accepted 9 June 2001
Key words: astrocytes, differentiation, neural stem cells, telomerase, TGF-β
Abstract
Serum-free mouse embryo (SFME) cells were established by D. Barnes et al., and are known to be a neural stem
cell line, which differentiate into astrocytes upon treatment with TGF-β. Therefore, SFME cells is thought to be a
model well suited to analyze the differentiation mechanism of neural stem cells. Until now, we have investigated the
regulation mechanisms of telomerase activity and telomere length in human cancer and normal cells. Telomerase
is the enzyme responsible for the synthesis and maintenance of telomere repeats located at chromosomal ends and
is normally expressed in embryonic and germline cells, but not in most normal cells. Here, using SFME cells, we
attempted to analyze the regulation mechanism of telomerase activity in neural stem cells and to detect a change
upon differentiation into astrocytes. When SFME cells were cultured in the presence of TGF-β, cells showed an
elongated morphology and decreased its growth to 50% of control culture. Cells also expressed the glial fibrillary
acidic protein (GFAP), a marker for astrocytes, indicating that TGF-βinduced differentiation in SFME cells from
neural stem cells into astrocytes. At the same time, TGF-βalso inhibited telomerase activity and repressed the
expression of the mouse telomerase reverse transcriptase (mTERT), demonstrating that SFME cells was vested
with a finite replicative life span upon treatment with TGF-β. To understand the mechanisms regulating mTERT
levels during differentiation into astrocytes, we have estimated the expression level of c-myc, which is known to
be a key molecule in activating the TERT promoter. As a result, TGF-β-treated SFME cells were shown to repress
the expression of c-myc. Furthermore, promoter analysis, using the 5-region of the mTERT gene, which possess
two E-box elements bound to c-Myc/Max, demonstrated that mTERT promoter activity greatly decreased in TGF-
β-treated SFME cells as compared to non-treated SFME cells. These suggest that c-myc might play a critical role
in the expression of mTERT, and that down-regulation of c-myc dependent upon the astrocytic differentiation in
SFME cells might cause the repression of mTERT in TGF-β-treated SFME cells.
Abbreviations: hTERT – human telomerase reverse transcriptase; mTERT – mouse telomerase reverse tran-
scriptase; BMP2 – bone morphogenetic protein 2; CNS – central nervous system; GAPDH – glyceraldehyde-
3-phosphate dehydrogenase; GFAP – glial fibrillary acidic protein; LIF – leukemia inhibitory factor; RT-PCR –
reverse transcription-polymerase chain reaction; SFME – serum-free mouse embryo; TGF-β– transforming growth
factor-β; TRAP – telomeric repeat amplification protocol.
Introduction
Telomeres are highly conserved sequences at the ends
of eukaryotic chromosome. In most mammalian cells,
telomeric DNA consists of tandem repeats of the
sequence TTAGGG. Telomeres are widely thought
to play a role in the maintenance of chromosome
structure and stability (Greider, 1996). Telomerase
138
is a ribonucleoprotein enzyme that synthesizes the
telomeric DNA sequence onto the 3ends of chro-
mosomes. Embryonic cells and germline cells have
been shown to express telomerase activity. In contrast,
most normal somatic cells express low or undetect-
able levels of telomerase, and as a consequence their
telomeres shorten with each cell division cycle (Har-
ley et al., 1990; Hastie et al., 1990), and eventually
reach the threshold length that leads to replicative sen-
escence as a result of genomic instability. On the other
hand, it has been suggested that multipotent cells, such
as embryonic and germline cells, acquire their immor-
tality by maintaining telomerase activity (Thomson et
al., 1998).
Multipotent stem cells would be an invaluable
source for regeneration studies of the injured or dam-
aged cells and organs. Recently, to regenerate and/or
rescue the damaged central nervous system (CNS),a
worldwide attention has been focused on the stud-
ies of neural stem cells. Neural stem cells exist not
only in the developing mammalian nervous system
but also within the adult nervous system of all mam-
malian organisms (McKay, 1997; Alvarez-Buylla et
al., 1998). Mouse embryonic neuroepithelial cells are
known to contain cells that have the potential to differ-
entiate into both neurons and glial cells like astrocytes
(Reynolds et al., 1992; Johe et al., 1996; Rajan and
McKay, 1998). Serum-free mouse embryo (SFME)
cells have been characterized as a multipotent neural
stem cell line. SFME cells cultured in the serum-
free medium continues to grow without reaching the
crisis stage or showing gross genomic alteration. Fur-
thermore, SFME cells do not show tumorigenicity in
syngeneic and athymic mice (Loo et al., 1987). In
addition, SFME cells differentiates into astrocytes in
the presence of TGF-βor serum (Sakai et al., 1990).
Therefore, SFME cells are thought to be valuable for
investigating the differentiation mechanism of neural
stem cells.
As mentioned above, SFME cells can be main-
tained for extended period, thus is thought to be
immortal, which suggests that these cells maintain its
immortality by telomerase. Previously, the total brain
of a murine embryo has been shown to express sig-
nificant level of telomerase activity (Greenberg et al.,
1998; Martin-Rivera et al., 1998). However, little is
known about the regulation of telomerase regulation
during brain development and neural differentiation.
In this report, we attempted to study the regulation
mechanisms of telomerase activity during the differen-
Figure 1. The effect of TGF-βon SFME cell proliferation. SFME
cells were cultured for 9 days in the presence of 10 ng ml1TGF-β
() or the normal growth medium (, control). Cells were counted
using a cell counter (Sysmex F-300, TOA, Kobe, Japan) every day.
tiation of neural stem cells into astrocytes using SFME
cells.
Materials and methods
Cells culture and reagents
SFME cells were cultured in Dulbecco’s modified
Eagle’s medium/F12 (Life Technologies, Gainthers-
burg, MD) supplemented with 100 µgml
1insulin
(Sigma, St. Louis, MO), 10 µgml
1transferrin
(Sigma), 10 µgml
1high-density lipoprotein (Bio-
medical Technologies Inc., Stoughton, MA), 50 ng
ml1EGF (Sigma), 25 µgml
1ampicillin (WAKO,
Osaka, Japan), 200 U ml1penicillin (Meiji, Tokyo,
Japan), 200 µgml
1streptomycin (Meiji) and 10 nM
sodium selenite (Sigma) in a humidified atmosphere
with 5% CO2at 37 C. SFME cells were treated for
the indicated periods of time with 10 ng ml1TGF-β
(AUSTRAL Biologicals, San Ramon, CA).
RT-PCR
Total RNA was prepared by using TRIzol reagent
(Life Technologies). 2.5 µg of total RNA was an-
nealed with 500 ng of oligo (dT)20 at 70 Cfor
10 min, followed by reverse transcription at 42 C
for 50 min using SuperScript II reverse transcriptase
(Life Technologies). PCR was performed by using
1µl of the RT-reaction mixture for the amplifica-
tion of either mTERT (mouse telomerase reverse tran-
scriptase), c-myc,GFAP (glial fibrillary acidic protein)
or mGAPDH in a total volume of 10 µl contain-
ing 0.25 units of Ta q polymerase (Roche Diagnostics,
139
Indianapolis, IN) and 10 pmol of primers spe-
cific for mTERT (5-TAGAGGATTGCCACTGGCTC-
3and 5-TCCAATGCTCTGCAGCTTGC-3), or for
c-myc (5-TTCTCTCCTTCCTCGGACTC-3and 5-
TGGCAGCTGGATAGTCCTTC-3), or for GFAP (5-
ATGATGGAGCTCAATGACCG-3and 5-GGTTT-
CATCTTGGAGCTTCT-3)orformGAPDH (5-
CCGTAGACAAAATGGTGAAGGT-3and 5-GTG-
GTGCAGGAGGCATTGCTGA-3). Amplifications
were performed 31 cycles (30 sec at 94 C, 30 sec
at 60 Cand1minat72C) for mTERT,27cycles
(30 sec at 94 C, 30 sec at 58 C and 1 min at 72 C)
for c-myc, 27 cycles (30 sec at 94 C, 30 sec at 55 C
and 1 min at 72 C) for GFAP and mGAPDH.All
PCR reactions were done under hot-start conditions
and analyzed by electrophoresis on 3% agarose gel.
Measurement of telomerase activity
Telomerase activity was measured by the PCR-based
TRAP (Telomeric Repeat Amplification Protocol) as-
say (Kim et al., 1994) with some modifications
(Katakura et al., 1997).
Primers and conditions for PCR were as previously
described (Katakura et al., 1997). Quantification of te-
lomerase activity was performed by the Kodak EDAS
system (Eastman Kodak Co., Rochester, NY).
Measurement of mTERT promoter activity
Genomic DNA was extracted from SFME cells us-
ing the DNA Extractor WB Kit (WAKO). Using this
mouse genomic DNA as template, the mTERT pro-
moter region (–1560 to –27) was isolated by PCR us-
ing specific primers (5-CCCCTCCCTCCTCTTCT-
TTG-3and 5-CACGTGCGGGAACCAAGATG-3)
as shown in Figure 5. This fragment containing the
mTERT promoter was inserted into Nhe I site of
the pGL3-Basic luciferase (firefly luciferase) plasmid
(Promega, Madison, WI). As a positive control, pGL3-
TK, which has the firefly gene under the transcription
control of TK promoter, was used. For promoter
assays, approximately 1 ×105cells/well in 24-well
plates were co-transfected with the mTERT promoter-
luciferase construct (300 ng/well) and the pRL-TK
(150 ng/well, Promega) which expresses the Renilla
luciferase and was used as an internal control for trans-
fection efficiency. All transfections were performed
using the LipofectAMINE PLUS Reagent (Life Tech-
nologies). In TGF-βtreated cells, cells were treated
with TGF-β(10ngml
1) after transfection. The cells
were harvested at 48 hr after transfection and the luci-
ferase assay was done using the Dual-Luciferase Re-
porter Assay System (Promega). Samples were coun-
ted for 10 sec in a LUMINESCENCE READER BRL-
301 (ALOKA, Tokyo, Japan), and the data is repres-
ented as the relative ratio of firefly luciferase/Renilla
luciferase light unit.
Results and discussions
TGF-βinduced differentiation of neural stem cells
into astrocytes
We used SFME cells, which is a cell line established
from 16-day-old Balb/c mouse embryo cells, to invest-
igate the change in telomerase activity during the dif-
ferentiation of neural stem cells. SFME cells is known
to retain neural stem cell phenotypes when cultured in
serum free media, and >80% of SFME cells differenti-
ate into astrocytes upon treatment with TGF-β(Sakai
et al., 1990). Here, we confirmed that differentiation
of SFME cells into astrocytes does indeed occur upon
treatment with TGF-β.TGF-βweakly inhibited the
growth of SFME cells (Figure 1), which was observed
from day 4 after treatment with TGF-β.
Furthermore, TGF-β-treated SFME cells showed
extended cell morphology characteristic to astrocytes
(Figure 2). RT-PCR analysis demonstrated that GFAP
was induced in TGF-β-treated SFME cells, but not in
non-treated SFME cells (Figure 2). These results sug-
gest that TGF-βinduced cell differentiation in SFME
cells to become astrocytes.
It has previously been shown that TGF-βinhib-
ited the growth of primary astrocytes (Toru-Delbauffe
et al., 1990; Vergelli et al., 1995), although TGF-
βinduced differentiated SFME cells continued to
grow at the reduced growth rate. It is unclear at
present why astrocytes differentiated from SFME cells
lacks the mechanisms of growth inhibition by TGF-
β. On the other hand, SFME cells were maintained
in serum-free medium supplemented with EGF and
other growth factors (Ernst et al., 1991). SFME cells
arrested growth at the G1 phase of cell cycle and sub-
sequently underwent apoptosis upon the withdrawal
of EGF (Rawson et al., 1990). Slinskey et al. (1999)
has reported that p53, p21 and MDM-2 are not re-
quired for this G1 arrest and apoptosis in SFME cells
by the withdrawal of EGF. According to these stud-
ies, p53 pathway in SFME cells does not appear to
mediate G1 arrest and apoptosis. It has been previ-
ously shown that TGF-βincreases the expression of
140
Figure 2. Effect of TGF-βon SFME cell differentiation. Photomicrographs of SFME cells grown for 2 days in the absence (A) or presence
of TGF-β(10ngml
1) (B). (C) Expression of GFAP mRNA during TGF-β-induced astrocytic differentiation. SFME cells were treated with
TGF-β(10ngml
1) for 2 days, and RT-PCR assays were performed to detect GFAP and GAPDH mRNA.
p21WAF1/CI P1, and thereby TGF-βinduces growth
suppression of several cell types (Pardali et al., 2000).
Sun et al. (1998) has reported that the overexpression
of MDM-2 in culture cells could overcome the anti-
proliferative effect of TGF-β. Therefore, incomplete
growth inhibition in response to TGF-βobserved in
differentiated SFME cells may be caused by defects in
p53 signaling pathway. Further studies are needed to
clarify the signaling pathway of TGF-βrelating to the
differentiation and proliferation of SFME cells.
Differentiated SFME cells into astrocytes lost its
telomerase activity
Several reports indicate that neuronal differentiation
leads to the down-regulation of telomerase activity
(Kurk et al., 1996; Chen et al., 1997; Fu et al., 1999,
2000; Tian et al., 1999; Haik et al., 2000). Here, we
also measured the telomerase activity of the TGF-β
induced differentiated SFME cells with the TRAP as-
say. Treatment of SFME cells with TGF-βat 10 ng
ml1resulted in a remarkable reduction of telomerase
activity, while non-treated SFME cells retained strong
telomerase activity (Figure 3), indicating that SFME
cells are immortal from the standpoint of a replicat-
ive life-span, and that SFME cells are vested with a
finite replicative life-span upon differentiation. There-
fore, the telomere length in differentiated SFME cells
would shorten during long-term culture because of
the loss of telomerase activity. These differentiated
cells would then enter into the senescence and crisis
stage. It has been demonstrated that some cell lines
suppressed telomerase activity during terminal differ-
entiation (Sharma et al., 1995; Albanell et al., 1996;
Bestilny et al., 1996; Savoysky et al., 1996), which
is distinct from our case in that SFME cells main-
tains its growth potential even after differentiation. We
can rephrase these results as that the suppression of
telomerase activity in SFME cells is dependent upon
differentiation into astrocytes, but not upon cell cycle
arrest. Suppression of telomerase activity independent
141
Figure 3. The effect of TGF-βon the telomerase activity of
SFME cells. SFME cells were subcultured in the absence or pres-
ence of TGF-β(10ngml
1). To measure telomerase activity
semi-quantitatively, 1 ×106cells were harvested, washed and lysed.
Telomerase activity was determined in each cell extract using the
TRAP assay method. The TRAP assay products were visualized by
SYBR Green I staining.
Figure 4. Expression of mTERT and c-myc mRNA during
TGF-β-induced astrocytic differentiation. SFME cells were treated
with TGF-β(10ngml
1) for 2 days, and RT-PCR assays were
performed to detect mTERT,c-myc and GAPDH mRNA.
of cell cycle arrest was also observed in our previous
study showing that TGF-βdecreased telomerase activ-
ity in human cancer cells with maintaining its growth
potential (Katakura et al., 1999). Both cases suggests
that TGF-βtransmits specific signals to suppress te-
lomerase activity both in cancer cells and neural stem
cells.
Undifferentiated SFME cells expressed high level
of telomerase activity (Figure 3, lane 1). Telomerase
activity has been shown to be responsible for the rep-
licative immortality of human cells, thus exists at high
level in germ lines, embryonic tissues and embryonic
stem (ES) cells (Wright et al., 1996; Thomson et
al., 1998). Therefore, the high level of telomerase
activity observed in SFME cells means that they have
an infinite replicative life-span. In fact, it has been
observed that SFME cells have been continuously pas-
saged without evidence of growth crisis or senescence
for over 200 doublings when cultured in serum-free
medium (Loo et al., 1987). Here, we conclude that
SFME cells are neuronal stem cells and might be a
useful model cell line for identifying the pathways or
regulation mechanisms of telomerase activity during
neuronal cell differentiation.
TGF-βrepressed the expression of mTERT and c-myc
mRNA
To elucidate the detailed mechanism for the down-
regulated telomerase activity in the differentiated
SFME cells, we next measured the expression level
of mTERT mRNA in the TGF-βtreated-SFME cells
using the RT-PCR analysis. mTERT expression was
shown to be repressed in SFME cells exposed to TGF-
βat 10 ng ml1for 2 days when compared with
the expression of the internal control gene GAPDH
(Figure 4). This result is consistent with the report
that expression of TERT was tightly linked to the en-
zymatic activity of telomerase (Greenberget al., 1998;
Martin-Rivera et al., 1998). Moreover, it has been re-
ported that mTERT mRNA levels parallel a marked
decrease in telomerase activity during terminal differ-
entiation of the mouse erythroleukemia cells (Green-
berg et al., 1998). Therefore, we conclude that the
down-regulated telomerase activity as shown in Fig-
ure 3 might be caused by the transcriptional repression
of mTERT mRNA. The regulation mechanisms of
mTERT expression must be elucidated to understand
the molecular events that lead to neural differenti-
ation. These findings suggest that the expression of
mTERT is also tightly associated with the differenti-
ation of SFME cells into astrocytes. However, very
little is known about the expression and regulation of
the mTERT gene during neural differentiation. It has
recently been reported that the repression of TERT
expression during differentiation was correlated with
the loss of c-myc expression (Armstrong et al., 2000).
In agreement with their report, the expression of c-
myc in SFME cells was also shown to decrease upon
differentiation by treatment with TGF-β(Figure 4).
These results suggested that the repression of c-myc
may trigger the repression of mTERT expression dur-
142
Figure 5. The effect of TGF-βon the transcriptional activity of mTERT promoter in SFME cells. A diagram of the mTERT promoter is shown
by fixing the ATG codon as +1 (A). Moreover, two E-box elements are shown in box (Greenberg et al., 1999). (B) The reporter constructs were
transfected into SFME cells, and then treatment with TGF-βwas done for 2 days. Two days after transfection, luciferase expression levels
for each reporter construct were assessed using the Dual-Luciferase Reporter Assay System (Promega). pGL3-Basic is a negative control, and
pGL3-TK is a positive control. Data for each reporter construct is shown as the mean and standard deviation for samples done in triplicate.
ing the neural differentiation process. As shown in
Figure 5A, the mTERT promoter possesses two E-box
elements bound to c-Myc/Max which are located at –
837 and –32 bp from the translation start point (+1).
Thus, we isolated mTERT promoter by PCR using
mouse genomic DNA as the template, then measured
the mTERT promoter activity in differentiated SFME
cells by luciferase assay. Figure 5B clearly demon-
strated that the mTERT promoter activity in TGF-β
induced differentiated SFME cells greatly decreased
as compared to non-differentiated SFME cells. Con-
sidering together with the fact that c-myc is known
to be a strong transcriptional activator for hTERT via
E-box elements (Horikawa et al., 1999; Takakura et
al., 1999; Wu et al., 1999), our results suggest that
down-regulation of c-myc may lead to the repres-
sion of mTERT promoter activity. Down-regulation
mechanisms of c-myc may be dependent upon neural
differentiation and/or be a response to the TGF-β
signaling. These questions must be clarified to elucid-
ate the mechanisms for mTERT repression dependent
upon neural differentiation.
Furthermore, mTERT expression in SFME cells
was not completely repressed when treated with TGF-
βfor 2 days (Figure 4). In addition, the growth
potential partially remained in TGF-βtreated-SFME
cells (Figure 1).
These results suggested that the entry of SFME
cells into a state of terminal differentiation might need
other various molecules than TGF-β, or the continu-
ously treatment of TGF-β. In fact, it has been reported
that there are multiple routes for differentiation of
CNS stem cells into astrocytes (Rajan and McKay,
1998). Nakashima et al. have demonstrated that LIF
and BMP2 function in synergy to induce differenti-
ation into astrocyte, but BMP2 or LIF alone did not in-
duce astrocyte development (Nakashima et al., 1999).
Thus, different growth factors may be necessary for
the complete neuronal differentiation of SFME cells.
Further studies on neural differentiation of SFME cells
may provide valuable information about the neuronal
aging, neuronal dysfunction and neuronal death, such
as Alzheimer’s disease and Parkinson’s disease.
References
Albanell J, Han W, Mellado B, Gunawardane R, Scher HI,
Dmitrovsky E & Moore MA (1996) Telomerase activity is
repressed during differentiation of maturation-sensitive but not
resistant human tumor cell lines. Cancer Res 56: 1503–1508.
143
Alvarez-Buylla A & Temple S (1998) Stem cells in the developing
and adult nervous system. J Neurobiol 36: 105–110.
Armstrong L, Lako M, Lincoln J, Cairns PM & Hole N (2000)
mTert expression correlates with telomerase activity during the
differentiation of murine embryonic stem cells. Mech Dev 97:
109–116.
Bestilny LJ, Brown CB, Miura Y, Robertson LD & Riabowol KT
(1996) Selective inhibition of telomerase activity during terminal
differentiation of immortal cell lines. Cancer Res 56: 3796–3802.
Chen AJ, Liao SK, Chow SE, Chen JK & Wang TCV (1997) Dif-
ferential inhibition of telomerase activity during induction of
differentiation in hematopoietic, melanoma, and glioma cells in
culture. Biochem Biophys Res Commun 237: 438–444.
Ernst T, Jackson C & Barnes D (1991) Karyotypic stability of
serum-free mouse embryo (SFME) cells. Cytotechnology 5:
211–222.
Fu W, Begley JG, Killen MW & Mattson MP (1999) Anti-apoptotic
role of telomerase in pheochromocytoma cells. J Biol Chem 274:
7264–7271.
Fu W, Killen M, Culmsee C, Dhar S, Pandita TK & Mattson
MP (2000) The catalytic subunit of telomerase is expressed in
developing brain neurons and serves a cell survival-promoting
function. J Mol Neurosci 14(1–2): 3–15.
Greenberg RA, Allsopp RC, Chin L, Morin GB & DePinho RA
(1998) Expression of mouse telomerase reverse transcriptase
during development, differentiation and proliferation. Oncogene
16: 1723–1730.
Greider CW (1996) Telomere length regulation. Annu Rev Biochem
65: 337–365.
Haik S, Gauthier LR, Granotier C, Peyrin JM, Lages CS, Dormont
D & Boussin FD (2000) Fibroblast growth factor 2 up regu-
lates telomerase activity in neural precursor cells. Oncogene 19:
2957–2966.
Harley CB, Futcher AB & Greider CW (1990) Telomeres shorten
during ageing of human fibroblasts. Nature 345: 458–460.
Hastie ND, Dempster M, Dunlop MG, Thompson AM, Green DK
& Allshire RC (1990) Telomere reduction in human colorectal
carcinoma and with ageing. Nature 346: 866–868.
Horikawa I, Cable PL, Afshari C & Barrett JC (1999) Cloning
and characterization of the promoter region of human telomerase
reverse transcriptase gene. Cancer Res 59: 826–830.
Johe KK, Hazel TG, Muller T, Dugich-Djordjevic MM & McKay
RDG (1996) Single factors direct the differentiation of stem cells
from the fetal and adult central nervous system. Genes Dev 10:
3129–3140.
Katakura Y, Yamamoto K, Miyake O, Yasuda T, Uehara N, Nakata
E, Kawamoto S & Shirahata S (1997) Bidirectional regulation
of telomerase activity in a subline derived from human lung
adenocarcinoma. Biochem Biophys Res Commun 237: 313–317.
Katakura Y, Nakata E, Miura T & Shirahata S (1999) Transforming
growth factor-βtriggers two independent-senescence programs
in cancer cells. Biochem Biophys Res Commun 255: 110–115.
Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho
PL, Coviello GM, Wright WE, Weinrich SL & Shay JW (1994)
Specific association of human telomerase activity with immortal
cells and cancer. Science 266: 2011–2015.
Kurk PA, Balajee AS, Rao KS & Bohr VA (1996) Telomere regula-
tion and telomerase inactivation during neuronal cell differenti-
ation. Biochem Biophys Res Commun 224: 487–492.
Loo DT, Fuquay JI, Rawson CL & Barnes DW (1987) Extended
culture of mouse embryo cells without senescence: Inhibition by
serum. Science 236: 200–202.
Martin-Rivera L, Herrera E, Albar JP & Blasco MA (1998) Expres-
sion of mouse telomerase catalytic subunit in embryos and adult
tissues. Proc Natl Acad Sci USA 95: 10471–10476.
McKay R (1997) Stem cells in the central nervous system. Science
276: 66–71.
Nakashima K, Yanagisawa M, Arakawa H, Kimura N, Hisatsune T,
Kawabata M, Miyazono K & Taga T (1999) Synergistic signal-
ing in fetal brain by STAT3-Smad1 complex bridged by p300.
Science 284: 479–482.
Pardali K, Kurisaki A, Moren A, ten Dijke P, Kardass is D & Mousta-
kas A (2000) Role of smad proteins and transcription factor Sp1
in p21Waf1/Cip1 regulation by transforming growth factor-β.J
Biol Chem 275: 29244–29256.
Rajan P & McKay RD (1998) Multiple routes to astrocytic differen-
tiation in the CNS. J Neurosci 18: 3620–3629.
Rawson C, Cosola-Smith C & Barnes D (1990) Death of serum-free
mouse embryo cells caused by epidermal growth factor depriva-
tion is prevented by cycloheximide, 12-O-tetradecanoylphorbol-
13-acetate, or vanadate. Exp Cell Res 186: 177–181.
Reynolds BA, Tetzlaff W & Weiss SJ (1992) A multipotent EGF-
responsive striatal embryonic progenitor cell produces neurons
and astrocytes. J Neurosci 12: 4565–4574.
Sakai Y, Rawson C, Lindburg, K & Barnes D (1990) Serum and
transforming growth factor-βregulate glial fibrillary acidic pro-
tein in serum-free-derived mouse embryo cells. Proc Natl Acad
Sci USA 87: 8378–8382.
Savoysky E, Yoshida K, Ohtomo T, Yamaguchi Y, Akamatsu K,
Yamazaki T, Yoshida S & Tsuchiya M (1996) Down-regulation
of telomerase activity is an early event in the differentiation of
HL60 cells. Biochem Biophys Res Commun 226: 329–334.
Sharma HW, Sokoloski JA, Perez JR, Maltese JY, Sartorelli AC,
Stein CA, Nichols G, Khaled Z, Telang NT & Narayanan
R (1995) Differentiation of immortal cells inhibits telomerase
activity. Proc Natl Acad Sci USA 92: 12343–12346.
Shay JW & Bacchetti S (1997) A survey of telomerase activity in
human cancer. Eur J Cancer 33: 787–791.
Slinskey A, Barnes D & Pipas JM (1999) Simian virus 40 large T
antigen J domain and Rb-binding motif are sufficient to block
apoptosis induced by growth factor withdrawal in a neural stem
cell line. J Virol 73: 6791–6799.
Sun P, Dong P, Dai K, Hannon GJ & Beach D (1998) p53-
independent role of MDM2 in TGF-β1 resistance. Science 282:
2270–2272.
Takakura M, Kyo S, Kanaya T, Hirano H, Takeda J, Yutsudo M
& Inoue M (1999) Cloning of human telomerase catalytic sub-
unit (hTERT) gene promoter and identification of proximal core
promoter sequences essential for transcriptional activation in
immortalized and cancer cells. Cancer Res 59: 551–557.
Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel
JJ, Marshall VS & Jones JM (1998) Embryonic stem cell lines
derived from human blastocysts. Science 282: 1145–1147.
Tian XX, Pang JC, To SS & Ng HK (1999) Restoration of wild-type
PTEN expression leads to apoptosis, induces differentiation, and
reduces telomerase activity in human glioma cells. J Neuropathol
Exp Neurol 58: 472-479.
Toru-Delbauffe D, Baghdassarian-Chalaye D, Gavaret JM, Courtin
F, Pomerance M & Pierre M (1990) Effects of transforming
growth factor-β1 on astroglial cells in culture. J Neurochem 54:
1056–1061.
Vergelli M, Mazzanti B, Ballerini C, Gran B, Amaducci L & Mas-
sacesi L (1995) Transforming growth factor-β1 inhibits the
proliferation of rat astrocytes induced by serum and growth
factors. J Neurosci Res 40: 127–133.
144
Wright WE, Piatyszek MA, Rainey WE, Byrd W & Shay JW (1996)
Telomerase activity in human germline and embryonic tissues
and cells. Dev Genet 18: 173–179.
Wu KJ, Grandori C, Amacker M, Simon-Vermot N, Polack A,
Lingner J & Dalla-Favera R (1999) Direct activation of TERT
transcription by c-MYC. Nat Genet 21: 220–224.
... Evidence for the role of TERT in ASCs is supported by the loss of this protein after differentiation. TERT expression is lost following differentiation of NSCs into astrocytes following TGF-B treatment [301] or NSCs into neurons following treatment with NGF [302,303]. ...
... For example, telomerase activity has been localized to cellular fractions containing Type A, B, and C neural progenitor cells (NPCs) from the adult mouse V-SVZ [269]. Differentiation of embryonic stem cells into astrocytes or neurons induces loss of telomerase activity [301,303]. As mice age, they exhibit decreased TERT expression in the V-SVZ, which is accompanied by reduced proliferation and neurogenesis [350], consistent with a decline in the stem cell pool. ...
Article
Telomerase reverse transcriptase (TERT) is expressed by quiescent adult stem cells (qASC) in numerous adult murine and human tissues but has never been explored in the adult brain. Here, these data demonstrate that TERT+ cells in the adult mouse brain represent a novel population of multipotent qASCs. TERT+ cells were localized to numerous classical neuro/gliogenic niches including the ventricular-subventricular zone, hypothalamus and olfactory bulb, as well as newly discovered regions of adult tissue plasticity such as the meninges and choroid plexus. TERT+ cells expressed neural stem cell markers such as Nestin and Sox2, but not markers of activated stem/progenitor cells, nor markers of mature neuronal or glial cells. TERT+ qASCs also rarely expressed the proliferation marker Ki67, further confirming a quiescent phenotype. When cultured, TERT+ cells behaved like brain stem cells by forming proliferative neurospheres. Lineage tracing of TERT+ cells in adult transgenic mice revealed large-scale expansion of TERT+ progeny and differentiation in multiple brain regions to diverse cell types. Lineage-traced cells expressed markers of mature neurons, oligodendrocytes, astrocytes, ependymal cells, microglia, and choroid epithelial cells, thus demonstrating the striking multipotency of this stem cell population in basal tissue turnover. Finally, the neurogenic treatment of caloric restriction (CR) in lineage tracing animals revealed a decrease in TERT-traced cell signal within the median eminence (ME) of the hypothalamus, with no change in the arcuate nucleus (ARC), when compared to unrestricted diet (UR)-treated animals. Single-cell RNA sequencing of TERT-traced cells in mice administered CR treatment also revealed an increase in the neuroprotective gene brain lipid binding protein (BLBP) in TERT-traced cells after 1 month of CR. As neuroprotection is a classical response to inflammation, we then studied the role of TERT+ cells in the inflammatory process of aging. TERT+ cell numbers varied with aging across neurogenic niches but remained a similar percentage of the full brain. However, TERT-traced cell signal increased significantly with aging, although label retention decreased. Together, these data demonstrate that TERT+ cells represent a new population of multipotent stem cells that contribute to basal brain plasticity and regeneration.
... This expression pattern indicates that telomerase is present in the brain during embryonic development, which is supported by its elevated expression in embryonic neuronal stem or progenitor cells . Additionally, NSCs lose telomerase activity upon differentiating into astrocytes or neurons (Kruk et al., 1996;Miura et al., 2001;Caporaso et al., 2003;Cheng et al., 2007). It remains unclear how exactly telomerase activity decreases in differentiating cells. ...
... NSCs possess the capability to self-renew and differentiate into mature nerve cells including neurons, astrocytes and oligodendrocytes (Miura et al., 2001;Ming and Song, 2011;Würth et al., 2014). The activity of telomerase rapidly decreases when NSCs stop dividing and differentiate into nerve cells (Kruk et al., 1996;Klapper et al., 2001). ...
Article
Full-text available
Telomerase, a specialized ribonucleoprotein enzyme complex, maintains telomere length at the 3′ end of chromosomes, and functions importantly in stem cells, cancer and aging. Telomerase exists in neural stem cells (NSCs) and neural progenitor cells (NPCs), at a high level in the developing and adult brains of humans and rodents. Increasing studies have demonstrated that telomerase in NSCs/NPCs plays important roles in cell proliferation, neuronal differentiation, neuronal survival and neuritogenesis. In addition, recent works have shown that telomerase reverse transcriptase (TERT) can protect newborn neurons from apoptosis and excitotoxicity. However, to date, the link between telomerase and diseases in the central nervous system (CNS) is not well reviewed. Here, we analyze the evidence and summarize the important roles of telomerase in the CNS. Understanding the roles of telomerase in the nervous system is not only important to gain further insight into the process of the neural cell life cycle but would also provide novel therapeutic applications in CNS diseases such as neurodegenerative condition, mood disorders, aging and other ailments.
... In the brain, telomerase activity is mainly restricted to zones with neural stem cells such as the subventricuar zone (SVZ) and dentate gyrus of the hippocampus. Adult neural stem cells possess telomerase activity which is downregulated during differentiation into neurons and astrocytes (Miura et al., 2001, Caporaso et al., 2003. Microglia cells as resident brain macrophages possess some telomerase activity in the adult brain which can also be transiently upregulated upon brain injury (Fu et al., 2002b). ...
Article
Full-text available
Telomerase, consisting of the protein subunit TERT and RNA component TERC, is best known for maintaining and extending human telomeres, the ends of linear chromosomes, in tissues, where it is active, such as stem cells, germline cells, lymphocytes and endothelial cells. This function is considered as canonical. However, various non-canonical functions for the protein part TERT have been discovered. There are multiple such roles which can interfere with several signalling pathways, cancer development and many other processes. One of these non-canonical functions includes shuttling of the TERT protein out of the nucleus upon increased oxidative stress into the cytoplasm and organelles such as mitochondria. Mitochondrial TERT is able to protect cells from oxidative stress, DNA damage and apoptosis although the exact mechanisms are incompletely understood. Recently, a protective role for TERT was described in brain neurons. Here TERT is able to counteract effects of toxic neurodegenerative proteins via changes in gene expression, activation of neurotrophic factors as well as activation of protein degrading pathways such as autophagy. Protein degradation processes are prominently involved in degrading toxic proteins in the brain like amyloid-β, pathological tau and α-synuclein that are responsible for various neurodegenerative diseases. These new findings can have implications for the development of novel treatment strategies for neurodegenerative diseases. The current review summarises our knowledge on the role of the telomerase protein TERT in brain function, in particular, under the aspect of age-related neurodegenerative diseases. It also describes various strategies to increase TERT levels in the brain.
... Telomere length has also been demonstrated to be important for neuronal differentiation and neuritogenesis [216] (see also review [217]). Its deficiency leads to a compromised olfactory bulb neurogenesis [215] although NSCs lose telomerase activity upon differentiation into astrocytes [218]. DPSCs also lose progressively their telomerase activity upon their spontaneous in vitro differentiation to osteoblastic/odontoblastic cells in conditions of high culture passages [212]. ...
Article
Full-text available
The conversion of healthy stem cells into cancer stem cells (CSCs) is believed to underlie tumor relapse after surgical removal and fuel tumor growth and invasiveness. CSCs often arise from the malignant transformation of resident multipotent stem cells, which are present in most human tissues. Some organs, such as the gut and the brain, can give rise to very aggressive types of cancers, contrary to the dental pulp, which is a tissue with a very remarkable resistance to oncogenesis. In this review, we focus on the similarities and differences between gut, brain and dental pulp stem cells and their related CSCs, placing a particular emphasis on both their shared and distinctive cell markers, including the expression of pluripotency core factors. We discuss some of their similarities and differences with regard to oncogenic signaling, telomerase activity and their intrinsic propensity to degenerate to CSCs. We also explore the characteristics of the events and mutations leading to malignant transformation in each case. Importantly, healthy dental pulp stem cells (DPSCs) share a great deal of features with many of the so far reported CSC phenotypes found in malignant neoplasms. However, there exist literally no reports about the contribution of DPSCs to malignant tumors. This raises the question about the particularities of the dental pulp and what specific barriers to malignancy might be present in the case of this tissue. These notable differences warrant further research to decipher the singular properties of DPSCs that make them resistant to transformation, and to unravel new therapeutic targets to treat deadly tumors. Keywords: alternative lengthening of telomeres; cancer stem cells; cell markers; colorectal cancer; dental pulp stem cells; glioma; pluripotency core factors; stem cells; telomerase.
... Mouse and human microglia due to their macrophage-related origin display some telomerase activity and TERT protein [30,28]. However, contradicting data exists for rodent astrocytes, with some studies demonstrating initial but over time subsiding telomerase activity during in vitro culture [30] while other studies found a decrease of telomerase activity and TERT expression during in vitro differentiation of mouse neural stem cells into astrocytes [31]. Our group did not detect TERT protein in astrocytes of the human hippocampus and cultured mouse astrocytes [28]. ...
... Therefore, BMVC can be used to distinguish between cancer cells and normal cells 25 . Similarly, stem cells also have unlimited proliferation ability 26 and can maintain their length of telomeres during process of differentiation [27][28] . Therefore, it is reasonable to assume that BMVC might be used to distinguish between stem cells and somatic cells. ...
Article
Full-text available
Up to now, there is no appropriate marker to specifically and effectively label living neural stem/precursor cells (NSPCs) to investigate NSPC migration and differentiation. Therefore, the purpose of this study was to develop a specific method for tracking NSPCs by a cell marker, 3, 6-bis (1-methyl-4-vinylpyridinium) carbazole diiodide (BMVC). It was found that the bright fluorescence spots could be rapidly observed in the nuclei of NSPCs, isolated from embryonic rat cerebral cortex, after incubation with BMVC for just 5 min and could be maintained for exceeding 7 days without obvious decay. Even NSPCs were induced to undergo differentiation; their daughter cells also expressed bright fluorescence spots. In contrast, foreskin fibroblasts, adipose-derived stem cells (ADSCs), bone marrow mesenchymal stem cells (BMMSCs), neurons, and glial cells did not express the bright fluorescence spot in the presence of BMVC. In addition, BMVC-labeled NSPCs could be tracked in wound healing and decelluarized rat brain models with minimal cell toxicity. Therefore, BMVC possessed specific function to distinguish between NSPCs and other stem and neuron-related cells. These results are very encouraging, since a cell marker favorable for long-term labeling NSPCs should be useful in the development of strategies for tracking the development of NSPCs in neuroscience research.
... It is known that cultured astrocytes under basal conditions exhibit a cyclical pattern of telomere lengthening and shortening and are able to divide for much longer periods of time than microglia (Flanary and Streit, 2004). Such replicative capacity is finite after the murine neural stem cell line has differentiated into astrocytes, because telomerase activity is limited and, with each cell division, telomeres shorten progressively (Miura et al., 2001). Moreover, other extratelomere functions are exerted by this cellular enzyme, including the regulation of calcium distribution (Lin et al., 2007), metabolism (Chung et al., 2005), growth factor secretion (Smith et al., 2003), mitochondrial function (Passos et al., 2007), energy balance (Bagheri et al., 2006), and apoptosis (Massard et al., 2006). ...
Article
Although HIV-associated neurocognitive disorders (HAND) result from injury and loss of neurons, productive infection routinely takes place in cells of macrophage lineage. In such a complex context, astrocytosis induced by local chemokines/cytokines is one of the hallmarks of HIV neuropathology. Whether this sustained astrocyte activation is able to alter telomere-aging process is unknown. We hypothesized that interaction of HIV with astrocytes may impact astrocyte telomerase activity (TA) and telomere length in a scenario of astrocytic activation measured by expression of glial fibrillary acidic protein (GFAP). To test this hypothesis, cultured murine astrocytes were challenged with pseudotyped HIV/vesicular stomatitis virus (HIV/VSV) to circumvent the absence of viral receptors; and GFAP, telomerase activity, and telomere length were quantified. As an early and transient event after HIV infection, both TA activity and telomere length were significantly augmented (P < 0.001). Later, a strong negative correlation (-0.8616, P < 0.0001) between virus production and telomerase activity was demonstrated. Once HIV production had reached a peak (7 dpi), the TA decreased, showing levels similar to those of noninfected cells. In contrast, the astrocyte became activated, exhibiting significantly increased levels of GFAP expression directly related to the level of HIV/VSV replication (P < 0.0001). Our results suggest that HIV-infected astrocytes exhibit early disturbance in their cellular functions, such as telomerase activity and telomere length, that may attenuate cell proliferation and enhance the astrocyte dysregulation, contributing to HIV neuropathogenesis. Understanding the mechanisms involved in HIV-mediated persistence by altering the telomere-related aging processes could aid in the development of therapeutic modalities for neurological complications of HIV infection. © 2013 Wiley Periodicals, Inc.
... Telomerase has been shown to play an indispensable role in the maintenance of neural stem cells (NSCs), while its deficiency results in the exhaustion of NSCs pool, leading to compromised olfactory bulb neurogenesis (Ferron et al. 2004). Telomerase expression in neural progenitors start to decline upon differentiation into neurons (Ferron et al. 2009), and correspondingly, neural stem cells loose telomerase activity upon differentiation into astroyctes (Miura et al. 2001). Interestingly, telomerase over-expression in neural cell lines inhibits neural differentiation (Richardson et al. 2007), suggesting that a fine balance in telomerase activity and telomere length is stringently regulated in neural stem cell compartment compared to other adult stem cell compartment such as BMSCs. ...
Article
Stem cell research holds a promise to treat and prevent age-related degenerative changes in humans. Literature is replete with studies showing that stem cell function declines with aging, especially in highly proliferative tissues/ organs. Among others, telomerase and telomere damage is one of the intrinsic physical instigators that drive agerelated degenerative changes. In this review we provide brief overview of telomerase-deficient aging affects in diverse stem cells populations. Furthermore, potential disease phenotypes associated with telomerase dysregulation in a specific stem cell population is also discussed in this review. Additionally, the role of telomerase in stem cell driven cancer is also briefly touched upon.
Article
Cell senescence is characterized by the irreversible arrest of cell proliferation and has been implicated as one of the critical causes of Alzheimer's disease, Parkinson's disease, and other neurodegenerative diseases. Telomere dysfunction, oxidative stress, DNA damage, senescence-associated secretory phenotype, and mitochondrial dysfunction contribute to the development of cellular senescence. Telomerase reverse transcriptase (TERT), which is the catalytic subunit of telomerase, can counteract cellular senescence with telomerase RNA template in a telomere-dependent manner. In addition, TERT has also been confirmed to exert extra-telomeric and neuroprotective roles in neurodegenerative diseases. In this review, we focus on the close relationship between cellular senescence and neurodegenerative diseases, and in particular, we elucidate the neuroprotective role of TERT in neurodegenerative diseases.
Chapter
In order to devise a novel yet simple method for evaluating the effect of environmental chemicals on nerve cell differentiation, we investigated the SFME system. This system is known to differentiate into astrocytes following induction with FBS or TGF-β. We used the RT-PCR method to detect the GFAP expression, an astrocyte specific marker, and determined the differentiation that accompanied the gene activation at transcription level. We then examined three environmental chemicals in order to determine whether they interfered with the GFAP gene activation. It was found that bisphenol A (BPA) and di-2-ethylhexyl phythalate (DEHP) suppressed the induction in a concentration-dependent manner, but that the effect of nonylphenol (NPL) was unclear. It was concluded that this induction system provides a simple and convenient way of evaluating the effect of environmental chemicals on nerve cell differentiation.
Article
Full-text available
Transforming growth factor–β (TGF-β) inhibits cell proliferation, and acquisition of TGF-β resistance has been linked to tumorigenesis. A genetic screen was performed to identify complementary DNAs that abrogated TGF-β sensitivity in mink lung epithelial cells. Ectopic expression of murine double minute 2 rescued TGF-β–induced growth arrest in a p53-independent manner by interference with retinoblastoma susceptibility gene product (Rb)/E2F function. In human breast tumor cells, increased MDM2 expression levels correlated with TGF-β resistance. Thus, MDM2 may confer TGF-β resistance in a subset of tumors and may promote tumorigenesis by interference with two independent tumor suppressors, p53 and Rb.
Article
Full-text available
The mitogenic actions of epidermal growth factor (EGF) were examined in low-density, dissociated cultures of embryonic day 14 mouse striatal primordia, under serum-free defined conditions. EGF induced the proliferation of single progenitor cells that began to divide between 5 and 7 d in vitro, and after 13 d in vitro had formed a cluster of undifferentiated cells that expressed nestin, an intermediate filament present in neuroepithelial stem cells. In the continued presence of EGF, cells migrated from the proliferating core and differentiated into neurons and astrocytes. The actions of EGF were mimicked by the homolog transforming growth factor alpha (TGF alpha), but not by NGF, basic fibroblast growth factor, platelet-derived growth factor, or TGF beta. In EGF-generated cultures, cells with neuronal morphology contained immunoreactivity for GABA, substance P, and methionine-enkephalin, three neurotransmitters of the adult striatum. Amplification of embryonic day 14 striatal mRNA by using reverse transcription/PCR revealed mRNAs for EGF, TGF alpha, and the EGF receptor. These findings suggest that EGF and/or TGF alpha may act on a multipotent progenitor cell in the striatum to generate both neurons and astrocytes.
Article
In the vertebrate central nervous system, multipotential cells have been identified in vitro and in vivo. Defined mitogens cause the proliferation of multipotential cells in vitro, the magnitude of which is sufficient to account for the number of cells in the brain. Factors that control the differentiation of fetal stem cells to neurons and glia have been defined in vitro, and multipotential cells with similar signaling logic can be cultured from the adult central nervous system. Transplanting cells to new sites emphasizes that neuroepithelial cells have the potential to integrate into many brain regions. These results focus attention on how information in external stimuli is translated into the number and types of differentiated cells in the brain. The development of therapies for the reconstruction of the diseased or injured brain will be guided by our understanding of the origin and stability of cell type in the central nervous system.
Article
: The effects of transforming growth factor β1 (TGFβ1) on DNA synthesis and functional differentiation of astroglial cells cultured in serum-free medium were investigated. TGFβ1 diminished and delayed the peak of DNA synthesis induced by serum. TGFβ1-treated cells were larger than control cells. This factor delayed the appearance of process-bearing cells induced by acidic fibroblast growth factor treatment and also affected the astrocyte-specific enzyme glutamine synthetase (GS), whose accumulation is under hydrocortisone (HC) control. TGFβ1 inhibited the induction of GS activity by HC in a dose- and time-dependent manner. Moreover, pretreatment with TGFβ1 for 4 h maintained the inhibition of GS activity for ˜16 h after removal of this factor from culture medium. These results suggest that TGFβ1 may be an important regulator of astrocyte growth and differentiation.
Article
Telomerase, the enzyme which maintains the ends of linear chromosomes in eukaryotic cells, is found at low levels in somatic stem cells but while this is incapable of preventing the progressive erosion of telomeres occurring as a consequence of cell division, such cells show greater proliferative capacity than normal somatic cells hence examination of telomerase activity in such stem cells is of interest. Our aim in this work was to examine the relationship between expression of the reverse transcriptase component (mTert) of murine telomerase. We report here the insertion of a reporter cassette comprising a segment of the promoter sequence of murine Tert gene coupled to the coding sequence of green fluorescent protein (GFP) into murine embryonic stem (ES) cells and show that this is sufficient for mimicking the expression of mTert. We show that the expression of mTert is very closely linked to telomerase activity and that both are substantially reduced upon differentiation of ES cells into more committed lineages giving us a potential reporter system for the selection and isolation of ES cells possessing different levels of telomerase activity.
Article
Mouse embryo cultures derived in serum-containing medium undergo growth crisis or senescence after fewer than 20 population doublings, followed by the emergence of genetically altered, polyploid 'immortalized' cells capable of growing indefinitely. Serum-free mouse embryo (SFME) cells, derived in medium in which serum is replaced with growth factors and other supplements, do not exhibit growth crisis or gross chromosomal aberrations when cultured for well over 100 population doublings and display other unique properties. We examined culture conditions and physiological factors affecting karyotypic stability in long term cultures of SFME cells derived from several mouse strains. Cloning SFME cells consistently isolated colonies with altered karyotype, even when the clones were derived from parent cultures with no karyotypic alterations. After 140-200 population doublings in vitro, the percentage of SFME cells showing hyperdiploidy or structural chromosomal abnormalities increased, although the modal chromosome number remained diploid. SFME cells transformed with molecularly cloned oncogenes did not show alterations in karyotype beyond that expected from the clonal origins of these cells, indicating that malignant transformation of SFME cells does not result in general karyotypic instability.
Article
Serum-free mouse embryo (SFME) cells, derived in medium in which serum is replaced with growth factors and other supplements, display distinctive properties: (i) SFME cells do not lose proliferative potential or show gross chromosomal aberration upon extended culture, (ii) these cells depend on epidermal growth factor for survival; and (iii) SFME cell proliferation is reversibly inhibited by serum. Treatment of SFME cells with serum or transforming growth factor beta led to the appearance of glial fibrillary acidic protein, a specific marker for astrocytes. The appearance of glial fibrillary acidic protein in cultures was reversed upon removal of transforming growth factor beta or serum. Cells with properties similar to SFME cells were also isolated from adult mouse brain. These results suggest a role for transforming growth factor beta in astrocyte differentiation in developing organisms and in response to injury and identify the cell type that has the unusual properties of SFME cells.