Calcitonin Receptor-Mediated Growth Suppression of HEK-293 Cells Is Accompanied by Induction of p21WAF1/CIP1 and G2/M Arrest
Andreas Evdokiou,
Liza-Jane Raggatt,
Gerald J. Atkins and
David M. Findlay
Department of Orthopaedics and Trauma University of
Adelaide The Royal Adelaide Hospital and the Hanson Centre
for Cancer Research at the Institute for Veterinary and Medical
Science Adelaide 5000, South Australia, Australia
 |
ABSTRACT
|
---|
We investigated the mechanisms by which calcitonin
(CT) suppresses cellular proliferation, using HEK-293 cells stably
transfected with either the rat C1a CT receptor (CTR) or the
insert-negative form of the human CTR. CT treatment of clonal cell
lines expressing either receptor type, but not untransfected HEK-293
cells, strongly suppressed cell growth in a concentration-dependent
manner. The reduction in cell growth with CT treatment could not be
attributed to cellular necrosis or apoptotic cell death, the latter
assessed by both DNA fragmentation analysis and caspase 3 (CPP-32)
assay. Growth inhibition was associated with an accumulation of cells
in the G2 phase of the cell cycle. CT treatment
of the human and rat CTR-expressing cell lines resulted in a rapid and
sustained induction of mRNA encoding the cyclin-dependent kinase
inhibitor, p21WAF1/CIP1, increased levels of
which were maintained at least 48 h after initiation of treatment.
Western blot analysis showed a rapid corresponding increase in
p21WAF1/CIP1 protein, whereas protein levels of
another member of the cyclin-dependent kinase inhibitor family,
p27kip1, were unchanged. In parallel with the
induction of p21, CT treatment reduced levels of p53 mRNA and protein.
CT treatment resulted in a specific cell cycle block in
G2, which was associated with inhibition of
Cdc2/cyclin B kinase activity as measured by histone H1
phosphorylation. There was no evidence for p21 association with this
complex despite the inhibition of Cdc2 activity. Evidence that p21
induction was causative of cell growth suppression was obtained from
p21 antisense oligonucleotide experiments. Treatment with a p21
antisense oligonucleotide blocked induction of p21 expression and
significantly reduced the CT-mediated growth inhibition. These
observations suggest that p21 is required for the
G2 arrest in response to CT, but argue against
a direct role of p21 in the inhibition of Cdc2 activity. These studies
suggest a novel regulation of cell cycle progression by CT and will
provide a basis for detailed examination of the molecular mechanisms
involved.
 |
INTRODUCTION
|
---|
Calcitonin (CT) is a 32-amino acid peptide hormone of thyroidal
origin, whose main recognized physiological role is the inhibition of
bone resorption by acting directly on osteoclasts (1). However, both CT
and its receptors (CTR) have also been identified in a large number of
other cell types and tissue sites (2), suggesting roles for the CT/CTR
system distinct from those involving calcium homeostasis. The CTR
belongs to a subclass of the large seven-transmembrane domain (7TMD) G
protein-coupled receptor (GPCR) family, which includes the receptors
for PTH/PTH-related peptide (PTHrP), glucagon, vasoactive intestinal
peptide, pituitary adenylate cyclase activating peptide, and
other peptide agonists (3). There is now growing evidence for an
important role for GPCRs in influencing cell growth and
differentiation, which is of particular relevance to development,
tissue remodeling and repair, and oncogenesis (4). Gains of function
mutations in receptors of this class have been shown to lead to
inappropriate cell growth. For example, Jansens metaphyseal
chondrodysplasia was recently shown to be due to constitutive activity
of the PTH/PTH-related peptide receptor, resulting in impairment of
growth and differentiation of chondrocytes in developing long bone
growth plates (5). In addition, inappropriate activity of GPCRs has
been implicated in carcinogenesis, with mutant TSH receptors, for
example, serving as definitive examples of oncoproteins for this
receptor class (6).
Multiple isoforms of the rat and human CTRs have been described, which
result from alternative splicing of the primary mRNA. In the case of
the rat CTR, two forms, termed C1a and C1b, differ structurally in that
C1b contains a 37-amino acid sequence in the second extracellular
domain, which is not present in the C1a form and which results in
different ligand binding properties (7). The human CTR (hCTR) is
primarily expressed as two functionally different isoforms, comprising
an insert-negative form and a form that contains 16 additional amino
acids inserted in the first intracellular loop (8, 9). Unlike the
insert-negative form, the insert-positive form does not signal via
phospholipase C/Ca2+-mediated pathways, although it retains
some signaling capacity via coupling to adenylate cyclase (8, 9). CTRs
are expressed in a number of human cancer cell lines, including those
of lung (10), prostate (11), and breast cancer origin (12, 13). In
addition, we have recently demonstrated the presence of mRNA encoding
the hCTR in all cases in a series of primary human breast tumors (14).
These results were of interest because of a longstanding observation by
Ng and co-workers (15), showing that CT treatment of T47D and MCF7
human breast cancer cells in culture potently reduced the proliferation
of these cells. Further evidence for a role for CT in cell growth
modulation was the more recent finding of a mitogenic action of CT in
human prostate cancer cells (11).
The mechanisms by which 7TMD receptors influence cell growth are not
well understood and are likely to be receptor- and cell type-specific.
Work to date has largely focused on the involvement of immediate
postreceptor signaling events (16, 17) and the modulation of
growth-related early response genes (18, 19). However, since
proliferation of eukaryotic cells depends on progression through the
cell cycle, proteins that exert control at cell cycle checkpoints are
likely to be important, as has been found in many other cellular
contexts. Increased levels of these proteins, which include the
cyclin-dependent kinase inhibitors (CKIs), are in general associated
with growth arrest and/or differentiation, although the particular
protein involved appears to be cell type dependent. Indeed, in
PTH-induced differentiation of osteoblasts, which was accompanied by an
inhibition of cell proliferation, a specific increase in the CKI,
p27Kip1, was associated with an accumulation of the number
of cells in G1 phase (20). Likewise, suppression of cell
growth is frequently found to be mediated by induction of the CKI,
p21WAF1/CIP1, by p53-dependent (21) or p53-independent
means (22). The aim of the present work was to elucidate the mechanisms
by which CT affects cell proliferation, using stably transfected
HEK-293 cell lines expressing single isoforms of the human or rat CTR.
The results suggest that CT inhibits progression from the
G2 phase of the cell cycle by mechanisms involving the
rapid and sustained induction of p21WAF1/CIP1 and
inhibition of Cdc2 kinase activity.
 |
RESULTS
|
---|
Cell Proliferation-Dose Response and Time Course
Our recent finding that the CTR gene is frequently expressed in
human primary breast cancer (14), and the longstanding demonstration of
the ability of CT to inhibit the growth of the CTR-bearing breast
cancer cell lines (15), prompted this study of the mechanisms of
CT-induced growth suppression. However, cloning of the CTR revealed
that the human and rat receptors exist in at least two functionally
different forms, arising by alternative splicing of the primary mRNA
transcript (8, 9), and we found that mRNA corresponding to both
receptor forms is expressed in T47D and MCF-7 cells. To avoid this
complexity, the experiments described here were performed in HEK-293
cells transfected with single isoforms of the rat or hCTR. HEK-293
cells were chosen because the functional characteristics of the CTR
have been carefully explored in this cell type (23, 24).
HEK-293 cells stably transfected with the C1a isoform of the rat CTR
(clone D11) were incubated with salmon CT (sCT) under conditions of
optimal mitogenic stimulus (10% FBS). A single addition of sCT
profoundly inhibited the proliferation of these cells in a
concentration- (Fig. 1A
) and time- (Fig. 2
) dependent manner. sCT did not
influence the growth of untransfected HEK-293 (Figs. 1A
and 2
). sCT
also reduced the growth rate of HEK-293 cells transfected with the
insert-negative isoform of the hCTR (HR12) (Figs. 1B
and 2
), suggesting
that the ability of the CTR to modulate cell growth is not species
dependent. Similar CT-induced growth suppression was also observed in a
number of independent HEK-293 clones expressing the human or rat CTR
(not shown). The growth suppression observed was dependent on receptor
activation, since the sCT[832] analog, which binds to rat and hCTRs
but does not elicit intracellular signaling (24), had no effect on the
rate of cell growth (Table 1
).
Interestingly, human CT (hCT), even at higher concentrations, had only
a minimal effect on the growth of D11 cells and a small, though
significant, effect on HR12 cells (Table 1
), despite being a potent
activator of adenylate cyclase at both the human and rat CTR (24). We
speculate that the difference between sCT and hCT, in terms of growth
inhibition, may relate to the different kinetics of receptor
interaction of the two ligands. Whereas sCT binds in a poorly
reversible manner to the CTR (10) and activates intracellular signaling
persistently, receptor-bound hCT is readily dissociable from CTRs
(25).

View larger version (19K):
[in this window]
[in a new window]
|
Figure 1. The Effect of CT Concentration on Cell
Proliferation
A, HEK-293 cells transfected with the rat C1a CTR (D11) (), or
untransfected HEK-293 cells ( ), were plated at 5 x
104 cells per well in 24-well plates. After 24 h, sCT
was added once at the indicated concentrations, and cells were
incubated for a further 72 h. B, HEK-293 cells transfected with
the hCTR (HR12) ( ) were treated with sCT as described in panel A.
Cells were harvested and counted on a hemocytometer. Each data point
indicates the mean ± SEM of triplicate
determinations. These results are representative of four experiments.
|
|

View larger version (18K):
[in this window]
[in a new window]
|
Figure 2. The Effect of CT on Cell Proliferation
HEK-293 ( ), D11 (), and HR12 ( ) cells were seeded at 2 x
104 cells per well in 24-well plates. Twenty four hours
after seeding cells remained untreated (open symbols) or
were treated with one addition of 10 nM sCT (closed
symbols). Cells were harvested and counted on a hemocytometer
24, 48, 72, and 96 h after sCT addition. Data points represent the
mean ± SEM of triplicate determinations, and the
results are representative of three experiments.
|
|
Apoptosis Analysis
Given the potent inhibition of cell proliferation produced by CT
treatment of CTR-transfected HEK-293 cells, we sought to determine
whether this might be mediated by increased cell death. Trypan blue
staining of CT- treated cells showed no difference in cell viability
between CT-treated and control cells. When the nuclei of CT-treated
cells were stained with 4',6-diamidino-2-phenylindole (DAPI) and
examined by confocal fluorescence microscopy, we found no evidence of
morphological changes typical of apoptosis, i.e. condensed
and fragmented nuclei. Consistent with these results, there was also no
indication of CT-induced apoptosis, as determined by caspase 3 (CPP-32)
assay (data not shown). When DNA was isolated and subjected to agarose
gel electrophoresis, no internucleosomal DNA fragmentation could be
detected, in either CT-treated or control cells (Fig. 3
). In contrast, DNA isolated from D11
cells expressing the rat CTR treated with sCT under low serum
conditions showed the characteristic DNA laddering indicative of
apoptosis (Fig. 3
). Similar results were obtained with cells expressing
the hCTR (data not shown). We conclude that CT does not influence cell
viability in HEK-293 cells under serum-replete conditions but may
increase the susceptibility of these cells to apoptotic death by a
variety of other stress signals including growth factor
deprivation.

View larger version (74K):
[in this window]
[in a new window]
|
Figure 3. Detection of DNA Fragmentation after Treatment with
CT
Parental HEK-293 and D11 cells were untreated or treated once with 10
nM sCT, hCT, or sCT[832]. DNA was isolated and
subjected to agarose gel electrophoresis. No internucleosomal DNA
fragmentation was observed when cells were grown in the presence of
10% serum. However, a low molecular-size DNA ladder, which is
characteristic of cells undergoing apoptosis, was observed in D11 cells
grown in the presence of sCT in 0.5% serum.
|
|
Cell Cycle Analysis
The lack of effect of CT on cell viability suggested that CT was
acting to inhibit passage of cells through the cell cycle. Analysis by
fluorescence-activated cell scanning (FACS) was performed to determine
the effect of CT on the distribution of cells in each phase of the cell
cycle. Treatment of D11 or HR12 cells with 10 nM sCT for
72 h elicited a prominent accumulation of cells in the
G2/M phase of the cell cycle and a concomitant decreased
proportion of cells in G1, compared with untreated cells
(Fig. 4
). Untransfected cells were not influenced by
CT treatment. These observations are consistent with the notion that CT
causes G2/M arrest. The time course of these changes is
shown in Fig. 5
, which indicates that
G2/M arrest was seen as early as 24 h after a single
addition of CT and that maximal effects on G2/M were seen
72 h after CT addition.

View larger version (25K):
[in this window]
[in a new window]
|
Figure 4. FACS Analysis of CT-Treated Cells
HEK-293, D11, and HR12 cells were untreated or treated with one
addition of sCT (10 nM). Cells were harvested 72 h
later, fixed, stained, and analyzed for DNA content, as described in
Materials and Methods. Shown is the distribution and
percentage of cells in the G1, S, and G2 phase
of the cell cycle, respectively.
|
|
CT Elevates Both p21 mRNA and Protein in CTR-Transfected Cells
To examine the mechanism(s) underlying CTR-mediated growth
suppression, the effects of CT on the expression of two members of the
Cip/Kip family of cyclin-dependent kinase (cdk) inhibitors,
p21Waf1/Cip1 and p27Kip1, were examined by
Northern blot and Western blot analysis. Treatment of D11 cells with
sCT resulted in a marked increase (
3 fold) in the levels of p21 mRNA
by 4 h of treatment. The levels of p21 mRNA increased with
increasing cell density in untransfected HEK-293 cells or D11 cells in
the absence of CT. A single addition of sCT resulted in a further
increase, which was sustained over the 48 h treatment period (Fig. 6
, A and B). Similar effects were seen in
HR12 cells, although with slightly different kinetics, so that p21 mRNA
levels did not peak until 24 h after treatment. There was no
effect of CT on p21 mRNA levels in untransfected HEK-293 cells. In
agreement with its failure to affect growth of CTR-expressing HEK
cells, sCT[832] did not influence p21 mRNA levels (data not
shown).

View larger version (38K):
[in this window]
[in a new window]
|
Figure 6. Expression of p21 and p53 mRNA after Treatment with
CT
Parental HEK-293 cells and cells stably transfected with either C1a
isoform of the rat CTR (clone D11) or the insert negative isoform of
the hCTR (clone HR12) were cultured for the times indicated in the
absence or presence of 10 nM sCT, added once. Total RNA was
extracted and Northern blot analysis performed. Panel A, 10 µg of
total RNA were electrophoresed through a 1% agarose formaldehyde gel,
transferred to nylon membrane, and hybridized with cDNA probes for p21
and p53 as indicated. Blots were rehybridized with a cDNA probe
specific for GAPDH to indicate RNA loading. Results were analyzed by
densitometry and expressed as a ratio of p21 mRNA/GAPDH mRNA (panel B)
and p53 mRNA/GAPDH mRNA (panel C).
|
|
To determine whether the increase of p21 mRNA was reflected in the
level of p21 protein, Western blot analysis was performed using a p21
monoclonal antibody. A corresponding large increase (
20 fold over
untreated cells) in the levels of p21 protein were detected in the D11
CTR transfectants within 4 h of CT treatment (Fig. 7A
). By 24 h of CT treatment, the
levels of p21 protein had declined but were consistently higher than in
untreated cells at this time. There was no effect of CT in
untransfected cells. In contrast to the observations with p21, CT
treatment had no appreciable effect on p27 protein levels, as
determined by Western blot analysis (Fig. 7B
).

View larger version (21K):
[in this window]
[in a new window]
|
Figure 7. Western Blot Analysis of p21 and p27 Protein after
Treatment with CT
Parental HEK-293 and D11 cells were untreated or treated once with 10
nM sCT for the times indicated. Total cell lysates were
prepared, and equal amounts of total cell protein (50 µg) were
separated by SDS-PAGE, transferred to PVDF membrane, and immunoblotted
with mouse monoclonal antibodies against p21 protein (panel A), p27
protein (panel B), and p53 protein (panel C). In each case, results
were analyzed by densitometry and expressed as bar charts.
|
|
Since the involvement of p21 in the control of cell growth is dependent
on p53 in some cell systems, steady state levels of p53 mRNA were
measured concomitantly with p21 mRNA. CT treatment resulted in a
consistent decrease in p53 mRNA levels in both D11 and HR12 cells (Fig. 6
, A and C). No change was seen in untransfected HEK-293 cells.
Similarly, the levels of p53 protein declined rapidly within 2 h
after treatment of HR12 cells with CT (Fig. 7C
).
The Effect of CT on Mitotic Index and p34cdc2 Kinase Activity
To investigate whether the CT-induced G2/M
accumulation was due to a specific block in G2 or in M
phase, the mitotic index of CT-treated cells was evaluated 24 h
after CT treatment. The number of mitotic cells was 10-fold lower in
CT-treated cells (3.6 ± 0.4) compared with controls (32.7 ±
5.1), indicative of a specific block in G2 and failure of
cells to enter mitosis. Because cyclin B1 protein levels reportedly
increase as cells approach M phase (26), we determined the level of
cyclin B1 in HR12 cells cultured in the presence or absence of sCT. As
shown in Fig. 8A
, culture of HR12 cells
in the presence of sCT resulted in a time-dependent increase in the
level of cyclin B1 protein, with a peak at 48 h after treatment,
coincident with the accumulation of cells in late G2 (Fig. 5
).

View larger version (27K):
[in this window]
[in a new window]
|
Figure 8. Effect of CT on Cyclin B1 Protein Levels and on
Cdc2 Protein Kinase Activity in HR12 Cells
HR12 cells were untreated or treated with sCT for different times, and
total cell extracts (50 µg) were assayed for cyclin B1 by Western
blotting (panel A). Aliquots of the same extracts were
immunoprecipitated with a p21 polyclonal antibody, and immune complexes
were resolved by electrophoresis and assayed for p21 (panel B). For
Cdc2 kinase activity, cells were treated with CT for the indicated
times or treated with the drug nocodazole for 24 h. Immune
complexes were resolved by electrophoresis and immunoblotted with Cdc2
(panel C, upper panel). The Cdc2 immunoprecipitated
complexes were assayed further for their ability to phosphorylate
histone H1 in vitro (panel C, lower
panel). For cyclin B1-associated kinase activity, extracts were
immunoprecipitated with a cyclin B1 polyclonal antibody, and kinase
activity was assessed as previously described (panel C, lower
panel). Phosphorylation of Cdc2 Tyr15 in total cell lysates (50
µg) treated with CT or nocodazole was detected by Western blotting
using a phospho-Cdc2 (Tyr15)-specific antibody (panel D).
|
|
A recognized explanation for G2 arrest is the inhibition of
Cdc2 kinase, an enzyme essential for the onset of mitosis in mammalian
cells. Inhibition of Cdc2 is a function of its phosphorylation status
(26) and association of p21 with the Cdc2/cyclin B complex (27). To
define further the kinetics of p21 up-regulation after CT treatment,
Western blot analyses of p21 immunoprecipitates isolated from HR12
cells were assayed for p21 protein. Figure 8B
shows that p21 was barely
detectable in immunoprecipitates of untreated cells but increased
significantly within 2 h after CT treatment, peaked at 4 h,
and remained markedly elevated in CT-treated cells when compared with
untreated cells. The induction of p21 before the onset of
G2 made it possible that p21 had associated with Cdc2 and
prevented its activation. However, when Cdc2 or cyclin B1
immunoprecipitates isolated from CT-treated HR12 cells were assayed for
the presence of p21 protein, we were unable to detect p21 associated
with Cdc2 or cyclin B1 at any time after CT treatment (data not shown).
In the converse experiment, Cdc2 was undetectable in the corresponding
p21 immunoprecipitates. To confirm that Cdc2 was successfully
immunoprecipitated from the lysates, Cdc2-immunoprecipitated complexes
were resolved by electrophoresis and assayed for Cdc2 by Western
blotting (Fig. 8C
, upper panel). Cdc2 was readily detectable
in the immune complexes, and the levels were not altered after
treatment with CT. To assess the kinase activity of Cdc2 at different
times after addition of CT, Cdc2 and cyclin B1 immunoprecipitates from
HR12 cells were analyzed for their ability to phosphorylate histone H1.
Cdc2- and and cyclin B1-associated kinase activity (Fig. 8C
) increased
transiently in the first 24 h after CT treatment but declined
significantly thereafter and was lowest at 48 and 72 h,
concomitant with accumulation of cells in G2. In contrast,
when cells were specifically arrested in mitosis with the
microtubule-depolymerizing drug, nocodazole, the histone H1 kinase
activity associated with Cdc2 was considerably higher than that in the
CT-treated cells (Fig. 8C
). To investigate the inactivation of Cdc2 by
CT on Tyr15 phosphorylation, cell extracts were immunoblotted with a
phospho-Cdc2Tyr15-specific antibody, which detects Cdc2 when
catalytically inactivated by phosphorylation at Tyr15. As shown in Fig. 8D
, CT treatment resulted in a modest but reproducible increase in
Cdc2Tyr 15 phosphorylation, which occurred at 48 and 72 h
posttreatment, consistent with inactive Cdc2 and G2 arrest.
In contrast, cells treated with nocodazole had unphosphorylated
Cdc2Tyr15. These data indicate that when M phase was blocked by
activation of the spindle microtubule-assembly checkpoint using
nocodazole, Cdc2 complexes were held in an active state manifested by
the lack of Cdc2 Tyr15 phosphorylation (Fig. 8D
). However, when cells
were arrested in G2 with CT, the kinase activity of Cdc2
complexes was low, and Cdc2 Tyr15 was maintained in the phosphorylated
and inactive state. Taken together, these results clearly demonstrate
that CT blocks progression into mitosis by inhibiting the kinase
activity of the Cdc2/cyclin B1. Since this effect appears to be
independent of p21, the exact role of p21 in the CT-mediated
G2 arrest remains to be determined.
Effect of Antisense p21 on Cell Growth after CT Treatment
The kinetics of p21 mRNA and protein up-regulation, peaking early
at 2 h after CT treatment, suggest that p21 participates in the
observed G2 arrest. To investigate whether p21 induction by
CT is a cause or a result of G2 arrest, we assessed
CT-induced growth inhibition in the presence of a p21 sense or
antisense oligonucleotide. As expected, incubation of
hCTR-expressing cells (HR12) with the sense oligonucleotide did not
influence the increase in p21 mRNA after CT treatment. However,
incubation with the p21 antisense oligonucleotide abolished the ability
of CT to increase p21 mRNA (Fig. 9A
) and
protein (Fig. 9B
). The effect of CT on cell growth in the presence of
the p21AS oligonucleotide was determined in four independent
experiments carried out in triplicate. As shown in Fig. 9C
, the p21AS
oligonucleotide significantly prevented the CT-mediated growth
inhibition, while the p21S oligonucleotide did not. There was no
significant difference between cell numbers in control, sense-, or
antisense oligonucleotide-treated cells, in the absence of CT, ruling
out nonspecific toxicity of the oligonucleotides. These results suggest
a causative role of p21 in CT-mediated growth inhibition.

View larger version (26K):
[in this window]
[in a new window]
|
Figure 9. Effect of Antisense p21 on the Growth of HR12 Cells
after CT Treatment
HR12 cells were untreated (-) or treated (+) with CT in the presence
or absence of either the sense or antisense p21 oligonucleotide.
Northern blot analysis was performed to assess the levels of p21 mRNA
at 24 and 48 h after treatment with CT (panel A). Cell extracts
were collected 48 h after CT treatment, and p21 protein levels
were assessed by Western blotting using a p21-specific antibody (panel
B). HR12 cells were trypsinized and counted 48 h post-CT treatment
(panel C). These results are representative of four independent
transfection experiments.
|
|
 |
DISCUSSION
|
---|
CT is best understood in its role as a potent negative regulator
of osteoclastic bone resorption (2). However, the more recent discovery
of CT production (2, 28), and expression of its receptor (2, 8), in
extraskeletal sites implies that CT may have actions unrelated to
calcium metabolism and protection of the skeleton. Relevant to the
present report is the longstanding finding of CT-induced growth
suppression in T47D and MCF7 human breast cancer cells (15). Together
with evidence for a mitogenic action of CT in a prostate cancer cell
line (11), these data draw attention to the specific ability of CT to
modulate cell growth and the more general involvement of the 7TMD class
of receptor in cell proliferation (4). The present study shows that sCT
treatment of HEK-293 cells stably transfected with the rat C1a CTR, or
the insert-negative form of the hCTR, potently decreased cellular
proliferation. Treatment with hCT, on the other hand, had a minimal
effect on the growth of D11 cells expressing the rat CTR and a
significant, but much lesser, effect than sCT on the growth of HR12
cells expressing the hCTR. There are several possible implications of
this finding. First, hCT may be antiproliferative only at the hCTR, as
seen also in T47D and MCF-7 breast cancer cells (15). In these
experiments, hCT was added repeatedly and inhibition of cell growth
occurred over a longer time course than the present experiment. Second,
the teleost or fish-like CTs may be more potent than hCT in terms of
growth inhibition. There is good evidence for the presence of fish-like
CTs in several mammalian tissues, including brain and pituitary (2),
where their physiology is not yet understood. Moreover, our results
suggest the possibility of using sCT therapeutically as an
antiproliferative agent in human cancer, given our finding of the
frequent expression of the CTR in primary breast tumors (14).
It may be that molecules in the CT family are involved physiologically
in cellular decisions regarding growth and differentiation, as has been
reported for several ligands for other members of the receptor subclass
to which the CTRs belong. For example, pituitary adenylate cyclase
activating peptide (PACAP) (16), vasoactive intestinal peptide
(17), and PTH (18) have also been shown to modulate cell growth, in
addition to their reported acute effects on differentiated cells. In
the case of PTH, there is in vitro (18, 29) and in
vivo (30) evidence for an important role in cell growth and
differentiation. PTH receptor deficiency (31) or constitutively active
receptor (5) lead to altered chondrocyte growth and differentiation,
manifested as deranged growth plates in long bones. Studies to date,
examining the mechanism by which this class of receptor alters cellular
growth, have largely focused on signaling events proximal to receptor
activation. The signaling pathways that regulate growth are complex and
apparently receptor- and cell-type specific (32). It was shown
previously that CT inhibition of T47D cell growth occurred in parallel
with a selective and sustained activation of the type II isoform of
protein kinase A (15). However, a more recent report of CT signaling in
HEK-293 cells showed Gq-mediated stimulation of Erk1/2 activity (23).
These results are significant in the present context because activation
of the mitogen-activated protein kinase pathway is known to correlate
with inhibition of cell proliferation (33). CT has been shown to
regulate the early response genes, c-fos and
c-jun, in several cell types (18, 19); however, the nuclear
actions of CT on cell growth have not previously been explored.
Inhibition of growth by CT in transfected HEK-293 cells was accompanied
by a rapid and prolonged induction of the cyclin-dependent kinase
inhibitor, p21Waf1/Cip1. P21Waf1/Cip1 has
largely been studied with respect to inhibition of cyclin-cdk complexes
involved in cell cycle arrest in the G1 phase of the cell
cycle, viz, cyclin D-cdk and cyclin E-cdk
(e.g. Refs. 22, 34). However, early experiments to assay
the activity of p21Waf1/Cip1 suggested that it was
potentially a universal inhibitor of cyclin kinases (35), and p21 mRNA
in human fibroblasts shows bimodal periodicity, with peaks in both
G1 and G2/M (36). Expression in the
G2/M phase suggests a role for p21Waf1/Cip1 in
the control of the cell cycle during this particular phase. In the
experiments reported here, we found that CT treatment of
CTR-transfected HEK-293 cells resulted in a prominent and prolonged
accumulation of cells in G2/M phase of the cell cycle, with
no detectable G1 arrest. Despite the significant
accumulation of cells with a G2/M DNA content, the
percentage of mitotic cells was 10-fold lower in CT-treated cells as
compared with controls, suggesting a specific inhibition of cells in
G2 and not in M. The results of the p21 antisense
oligonucleotide experiments suggest that p21Waf1/Cip1 is
causally involved in the CT-mediated growth suppression in HEK-293
cells. In addition, the early elevation of p21Waf1/Cip1,
preceding inhibition of cell growth and cell cycle progression, and its
sustained high levels in CT-treated cells, is consistent with the
activity of this protein in many other cellular systems in which growth
inhibition is observed. However, the role of p21 at the G2
checkpoint remains to be clarified. We obtained no evidence for the
presence of p21 protein in the Cdc2/cyclin B1 complex despite
inhibition of Cdc2 kinase activity, arguing against a direct role for
p21 in inhibiting Cdc2 kinase. Thus the significance of a sustained
induction of p21 and the molecular mechanisms underlying the observed
cell cycle arrest in G2 after CT treatment remain to be
clarified.
The association of Cdc2 kinase with its cyclin partners has been shown
to play an important role during G2 to M progression in
mammalian cells (37). The activity of Cdc2 is regulated positively by
phosphorylation of threonine 161 (38) and negatively by
phosphorylation on Tyr-15 and Thr-14. Inhibitory phosphorylation of
Cdc2 on residues Tyr-15 and Thr-14 must be removed by the action of the
dual specificity phosphatase Cdc25 for the Cdc2/cyclin B complex to
become active as the driving force for mitosis (26). Here, we have
shown that CT induces a specific block in G2 phase of the
cell cycle by a mechanism involving the inhibition of Cdc2 kinase
activity. Interestingly, the activity of Cdc2 increased transiently in
the first 24 h after CT treatment but declined significantly
thereafter and was lowest at 48 and 72 h, concomitant with cells
accumulating in G2. This transient increase in Cdc2 kinase
activity was reproducibly observed and its significance will require
further investigation.
The promoter for the p21Waf1/Cip1 gene contains multiple
p53-response elements (39) and induction of p21Waf1/Cip1
has been shown to occur under p53-dependent conditions (40, 41).
However, induction in other situations appears not to require p53 (36).
In the CTR transfectants, CT induction of p21Waf1/Cip1 was
unlikely to have been p53-mediated since p53 mRNA levels were regulated
inversely to p21Waf1/Cip1 mRNA levels. The explanation for
this, and the possible involvement of reduced p53 levels in CT-induced
growth suppression, remains to be investigated. The reduced expression
of p53 correlated with the arrest of CT-treated cells in
G2, rather than G1, which is more typically the
case when p21Waf1/Cip1 induction is secondary to
up-regulation of p53 (reviewed in Ref. 42). In addition, up-regulation
of p53 in some cell types is associated with the induction of apoptotic
death (reviewed in Ref. 42), and the decrease in proliferation of
CT-treated HEK-293 cells in serum-replete medium clearly did not
precede apoptosis. It is interesting to speculate that the observed
decrease in p53 mRNA and protein levels might represent a
mechanism to protect the CT-treated cells from apoptosis after
G2 arrest.
These studies suggest a novel regulation of cell cycle progression by
CT and provide the basis for further detailed examination of the
molecular mechanisms linking activation of the CTR to proliferation.
The recent implication of CT/CTR in human cancers (11, 14) and early
embryonic development (28, 43) serve to further stimulate this line of
enquiry.
 |
MATERIALS AND METHODS
|
---|
Cell Culture
Human embryonic kidney (HEK-293) cells were maintained in DMEM
supplemented with 10% heat-inactivated FBS, 2 mM
glutamine, 100 U/ml penicillin, and 0.1 mg/ml streptomycin, as
previously reported (23). Stably transfected HEK-293 cells were
maintained in 200 µg/ml G418, which was removed before commencement
of experiments. Cells were grown at 37 C in a humidified atmosphere
with 5% CO2.
Stable Transfection of HEK-293 Cells
Two cell lines, D11 and F12, which express the rat C1a isoform
of the rat CTR in HEK-293 cells, have been described previously (23).
To compare the growth effects of the hCTR in the same cell type,
HEK-293 cells were transfected with the insert-negative isoform of the
hCTR. The hCTR cDNA was ligated into the mammalian expression vector
Zem228CC (both cDNA and vector were gifts from Zymogenetics Inc,
Seattle, WA). Using a modified calcium phosphate transfection method
(44), cells were transfected with 10 µg plasmid DNA/flask, together
with 20 µg of inert herring sperm DNA (Sigma Chemical Co., St Louis, MO), in 25-cm2 culture flasks at a
cell density of approximately 50%. Twenty-four hours after
transfection, neomycin selection was commenced with the addition of 400
µg/ml G418 (Life Technologies, Inc., Glen Waverley,
Victoria, Australia) and maintained for 2 weeks. Neomycin-resistant
colonies were picked manually, propagated in the continual presence of
G418 (200 µg/ml), and screened for binding of
[125I]sCT. A number of clones were obtained with
different levels of receptor expression. The cell line designated HR12,
which expressed 14.3 ± 3.7 x 105 receptors per
cell (mean ± SEM, n = 3), and bound
[125I]sCT with a Kd of 2.0 ± 0.4
nM (mean ± SEM, n = 3), was
primarily used for this study. As described for the C1a rat CTR, CT
treatment of HR12 cells also resulted in increased intracellular CAMP
levels and intracellular Ca2+ fluxes. Several other
clones expressing the hCTR yielded similar results, with respect to
cell growth and induction of p21WAF1/CIP1.
Cell Growth Analysis
To determine the effect of CT concentration on cell
proliferation, cells were seeded at 5 x 104 cells per
well in 24-well plates and incubated for 72 h in the presence of
increasing concentrations (10-11
M10-7 M) of sCT, hCT, or the
832 analog of sCT (Peninsula Laboratories, Inc. Belmont,
CA). To determine the effect of sCT on the rate of cell growth, cells
were plated in 24-well plates at 2 x 104 cells per
well in standard culture medium in the presence or absence of 1 x
10-8 M sCT for 14 days. Cells were harvested
by trypsinization and were counted manually in triplicate using a
hemocytometer.
Apoptosis Analysis
DNA Fragmentation
Cells were harvested by washing twice with PBS, after 72 h in the
presence or absence of 10 nM sCT, and incubated overnight
at 37 C in lysis buffer containing 10 mM Tris, pH 8.0, 5
mM EDTA, 100 mM NaCl, 1.0% SDS, and 200
µg/ml proteinase K. DNA was extracted twice with equal volumes of
phenol-chloroform-isoamylalcohol (25:24:1) and then precipitated in
ethanol. Samples were electrophoresed in a 1.2% agarose gel, stained
with ethidium bromide, and visualized under UV light.
Caspase 3 Assay
CPP-32/caspase 3 protease activity was assayed by the cleavage of
DEVD-AFC, a fluorogenic substrate based on the peptide sequence of the
caspase-3 cleavage site of poly(ADP-ribose) polymerase. Cells were
cultured for 48 h in the presence or absence of 10 nM
sCT, washed in PBS, and resuspended in NP-40 lysis buffer. To each tube
containing 8 µM of substrate in 1 ml of protease buffer
(50 mM HEPES, 10% sucrose, 10 mM
dithiothreitol, and 0.1%
3-[(chloramidopropyl)dimethylamino]-1-propanesulfonate,
pH 7.4), was added 2040 µl of cell lysate, and the mixture was
incubated at room temperature for 45 min. Caspase activity was
quantitated by measurement of yellow-green fluorescence at 505 nm
(excitation at 400 nm), due to release of AFC, in a LS50
spectrofluorimeter (Perkin Elmer Corp., Norwalk,
CT).
Cell Cycle Analysis
Cells were removed from culture dishes by trypsinization, collected by
centrifugation, resuspended in ice-cold PBS, and then fixed in absolute
methanol for at least 30 min. Cells were then washed in PBS containing
0.5% Tween 20, followed by two washes in PBS containing 2% FCS, and
then cells were resuspended in PBS/2% FCS containing 40 µg/ml RNase
A and incubated for 20 min at 37 C. Cells were washed in PBS/2% FCS
and were finally resuspended in PBS containing propidium iodide at a
final concentration of 20 µg/ml. The stained nuclei were analyzed
using a flow cytometer (Epics Profile, Coulter Corp., Hialeah,
FL). Cell cycle distribution was based on 2 N and 4
N DNA content.
Mitotic Index Analysis
Cells treated or not with 10 nM sCT for 24 h
were trypsinized, collected by centrifugation, washed in PBS, and
treated with hypotonic solution of 0.075 M KCl at 37 C for
30 min. After centrifugation, cells were fixed by the dropwise addition
of freshly prepared fixative (methanol-acetic acid, 3:1),
recentrifuged, and resuspended in fixative. Cells were dropped onto
clean microscope slides and stained with a 1:25 dilution of Giemsa
(Sigma Chemical Co.) in PBS for 30 min. Mitotic indices
were calculated as the number of cells with condensed chromosomes with
at least 3000 cells being examined from 10 different fields of
view.
RNA Extraction and Northern Blot Analysis
For Northern blot analysis, cells were seeded at a density of
1 x 105 cells per 25-cm2 flask, allowed
to attach for 2 days, and incubated for 4, 24, and 48 h in the
presence or absence of 10 nM sCT. Total RNA was isolated at
the indicated times using the TRIZOL Reagent (Life Technologies, Inc.) according to the manufacturers instructions. Total RNA
(10 µg per lane) was electrophoresed in formaldehyde/1% agarose
gels, transferred to Hybond N+ nylon membranes
(Amersham Pharmacia Biotech, Castle Hill, New South Wales,
Australia), and immobilized by UV cross-linking. Membranes were
prehybridized for 3 h at 42 C in 1 M NaCl, 1% SDS,
10% dextran sulfate, 50% formamide, and 100 µg/ml of heat-denatured
herring sperm DNA and hybridized with p21WAF1/CIP1 cDNA (a
kind gift from Dr. Helena Richardson, The University of Adelaide, South
Australia, Australia), or p53 cDNA (a kind gift from Dr. Roger Reddel,
Childrens Medical Research Institute, Westmead, New South Wales,
Australia) radiolabeled with [
-32P]dCTP by random
priming using the Giga prime kit (Bresatec, South Australia). To allow
quantitation of mRNA signals, the same filters were stripped and then
reprobed with a 450-bp 32P-labeled PCR-generated DNA
fragment of human glyceraldehyde-3-phosphate dehydrogenase (GAPDH).
Northern blots were analyzed using the PhosphorImager SF
(Molecular Dynamics, Inc., Sunnyvale, CA).
Western Blotting
Cells were lysed in lysis buffer containing 10 mM
Tris HCl, pH 7.6, 150 mM NaCl, 1% Triton X-100, 0.1% SDS,
and stored at -70 C until ready to use. Cell extracts were mixed with
an equal volume of sample buffer containing 12 mM Tris-HCl,
pH 6.8, 6% SDS, 10% ß- mercaptoethanol, 20% glycerol and
0.03% bromophenol blue. Protein samples were boiled for 5 min
and electrophoresed under reducing conditions in 14% polyacrylamide
gels. Separated proteins were electrophoretically transferred to PVDF
transfer membrane (NEN Life Science Products,
Boston, MA) and blocked in PBS containing 5% blocking reagent
(Amersham Pharmacia Biotech, Piscataway, NJ) for 1 h
at room temperature. Immunodetection was performed overnight at 4 C in
PBS/blocking reagent containing 0.1% Tween 20, using mouse monoclonal
antibodies to p21 (C24420), p27 (K25020) (Transduction Laboratories, Inc., Lexington, KY), p53 (clone PAB1801)
(Zymed Laboratories, Inc., South San Francisco, CA) and
rabbit polyclonal antibodies to cyclin B1 (H-433) and Cdc2 (C-19)
(Santa Cruz Biotechnology, Inc., Santa Cruz, CA). Filters
were rinsed several times with PBS containing 0.1% Tween 20 and
incubated with 1:5000 dilution of antimouse or antirabbit alkaline
phosphatase-conjugate (Amersham Pharmacia Biotech,
Arlington Heights, IL) for 1 h. Bound proteins were detected and
quantitated using the Vistra ECF substrate reagent kit (Amersham Pharmacia Biotech) on a Fluorimager (Molecular Dynamics, Inc.).
Immunoprecipitation and Kinase Activity
For immunoprecipitation as for kinase activity determination,
rabbit polyclonal antibodies to p34cdc2, p21, or cyclin B1
(Santa Cruz Biotechnology, Inc.) were attached to protein
A-Sepharose (Pharmacia Biotech) by incubation at 4 C for
24 h. Antibody-attached Sepharose was then added to 300 µg of
precleared cell lysates and incubated for a further 24 h at 4 C on
a rotating platform. Immune complexes were recovered and washed three
times with lysis buffer. For immunoprecipitation, immune complexes were
separated on 14% SDS-polyacrylamide gels as described above, and a
mouse monoclonal p21 antibody or rabbit polyclonal p34cdc2
was used for immunoblot analysis. For p34cdc2 kinase
activity, immune complexes were washed three times with kinase assay
buffer (50 mM Tris-HCl, pH 7.4, 10 mM
MgCl2, 1 mM dithiothreitol). Phosphorylation of
histone H1 was measured by incubating the beads with 40 µl of a
reaction mixture containing 25 µM ATP, 2 µg histone H1
(Roche Molecular Biochemicals, Nutley, NJ) and 2.5 µCi
[32P]
-ATP (3000 Ci/mmol; Bresatec) in kinase assay
buffer for 30 min at 37 C. The reaction was stopped by boiling the
sample in 2 x SDS sample buffer for 5 min, and samples were
resolved by SDS-PAGE. Gels were analyzed using the PhosphorImager SF
(Molecular Dynamics, Inc.).
Antisense Oligonucleotides
Phosphorothioate oligodeoxynucleotides (2 µg/ml) and
lipofectin (Life Technologies, Inc.) (10 µg/ml) were
incubated in serum free DMEM for 45 min at room temperature. The
oligonucleotide-lipofectin mixture was diluted with serum-containing
medium and added to the cells. After 4 h of preincubation with the
oligonucleotide mixture, cells were treated with 10 nM sCT
for up to 72 h. To confirm that the antisense oligonucleotide had
the desired effect of blocking p21 up-regulation by sCT, p21 mRNA was
monitored by Northern and Western blotting at 24 and 48 h after
treatment with sCT. At the indicated times, cells were trypsinized from
plates and counted. The antisense oligonucleotide was based on the p21
coding sequence p21AS, which is complementary to the region of the
initiation codon (5'-TCC CCA GCC GGT TCT GAC AT-3') and the sense p21
(Sp21) as control (5'-ATG TCA GAA CCG GCT GGG GA-3'). These
oligonucleotides were purchased from Life Technologies, Inc.
 |
ACKNOWLEDGMENTS
|
---|
The authors are grateful for reagents and advice from
Zymogenetics Inc. (Seattle, WA) and Dr. Prue Cowled and Lefta Leonardos
at the Queen Elizabeth Hospital (Adelaide, South Australia, Australia).
We are also grateful to Dr Matthew OConnell at the Peter MacCallum
Cancer Institute (Melbourne, Victoria, Australia) for useful
discussions.
 |
FOOTNOTES
|
---|
Address requests for reprints to: Associate Professor David M. Findlay, Department of Orthopaedics and Trauma, Level 4, Bice Building, Royal Adelaide Hospital, North Terrace, Adelaide 5000, South Australia, Australia.
This work was supported by The Kathleen Cunningham Foundation for
Breast Cancer Research, Australia, and grants from the Adelaide
Bone and Joint Research Foundation and Bristol-Myers Squibb Co./Zimmer. L.-J. Raggatt was supported by the Australian
Research Council Small Grants scheme.
Received for publication December 10, 1998.
Revision received June 30, 1999.
Accepted for publication July 8, 1999.
 |
REFERENCES
|
---|
-
Chambers TJ, Magnus CJ 1982 Calcitonin alters behaviour
of isolated osteoclasts. J Pathol 136:2739[Medline]
-
Martin TJ, Findlay DM, Moseley JM Sexton PM 1988 Calcitonin.
In: Alvioli LV, Krane SM (eds) Metabolic Bone Disease and Clinically
Related Disorder. Academic Press, San Diego, CA, pp 95121
-
Sprengel R, Eva C 1994 Hormone receptors. In: Peroutka JS
(ed) Handbook of Receptors and Channels. CRC Press, Boca Raton, FL, pp
153157
-
Dhanasekaran N, Heasley LE, Johnson GL 1995 G protein-coupled
receptor systems involved in cell growth and oncogenesis. Endocr Rev 16:259270[Medline]
-
Schipani E, Kruse K, Juppner H 1995 A constitutively active
mutant PTH-PTHrP receptor in Jansen-type metaphyseal
chondrodysplasia. Science 268:98100[Medline]
-
Parma J, Duprey L, Van Sande J, Cochaux P, Gervy C, Mochel J,
Dumont J, Vassart G 1993 Somatic mutations in the thyrotropin receptor
gene cause hyperfunctioning thyroid adenomas. Nature 365:649651[CrossRef][Medline]
-
Sexton PM, Houssami S, Hilton JM, OKeeffe LM, Center RJ,
Gillespie MT, Darcy P, Findlay DM 1993 Identification of brain isoforms
of the rat calcitonin receptor. Mol Endocrinol 7:815821[Abstract]
-
Kuestner RE, Elrod R, Grant FJ, Hagen FS, Kuijper JL,
Matthews SL, Sheppard PO, Stroop SD, Thompson DL, Whitmore T, Findlay
DM, Houssami S, Sexton PM, Moore, EE 1994 Cloning and characterization
of an abundant subtype of the human calcitonin receptor. Mol Pharmacol 46:246255[Abstract]
-
Moore EE, Kuestner RE, Stroop SD, Grant FJ, Matthewes SL,
Brady CL, Sexton PM, Findlay DM 1995 Functionally different isoforms of
the human calcitonin receptor result from alternative splicing of the
gene transcript. Mol Endocrinol 9:959968[Abstract]
-
Findlay DM, DeLuise M, Michelangeli VP, Ellison M, Martin TJ 1980 Properties of a calcitonin receptor and adenylate cyclase in BEN
cells, a human cancer cell line. Cancer Res 40:13111317[Abstract]
-
Shah GV, Rayford W, Noble MJ, Austenfeld M, Weigel J, Vamos S,
Mebust WK 1994 Calcitonin stimulates growth of human prostate cancer
cells through receptor-mediated increase in cyclic adenosine
3',5'-monophosphates and cytoplasmic Ca2+ transients.
Endocrinology 134:596602[Abstract]
-
Findlay DM, Michelangeli VP, Moseley JM, Martin TJ 1980 Calcitonin binding and degradation in two cultured human breast cancer
lines (MCF-7 and T47D). Biochem J 196:513520
-
Findlay DM, Michelangeli VP, Eisman JA, Frampton RJ, Moseley
JM, MacIntyre I, Whitehead R, Martin TJ 1980 Calcitonin and
1,25-dihydroxyvitamin D receptors in human breast cancer cell lines.
Cancer Res 40:47644767[Abstract]
-
Gillespie MT, Thomas RJ, Pu Z-Y, Zhou H, Martin TJ, Findlay,
DM 1997 Calcitonin receptors, bone sialoprotein and osteopontin are
expressed in primary breast cancers. Int J Cancer 73:812815[CrossRef][Medline]
-
Ng KW, Livesey SA, Larkins RG, Martin TJ 1983 Calcitonin
effects on growth and on selective activation of type II isoenzyme of
cyclic adenosine 3':5'-monophosphate-dependent protein kinase in T47D
human breast cancer cells. Cancer Res 43:794800[Abstract]
-
Morisset J, Douziech N, Rydzewska G, Buscail L, Rivard N 1995 Cell signalling pathway involved in PACAP-induced AR42J cell
proliferation. Cell Signal 7:195205[CrossRef][Medline]
-
Kimata H, Yoshida A, Ishioka C, Fujimoto M, Furusho K 1996 Vasoactive intestinal peptide enhances immunoglobulin production and
growth in human plasma cells via mechanisms that may involve protein
kinase C. J Clin Endocrinol Metab 81:30243032[Abstract]
-
Lacroix M, Body JJ 1997 Regulation of c-fos and c-jun
expression by calcitonin in human breast cancer cells. Calcif Tissue
Int 60:513519[CrossRef][Medline]
-
Kobayashi T, Sugimoto T, Saijoh K, Fukase M, Chihara K 1994 Calcitonin directly acts on mouse osteoblastic MC3T3E1 cells to
stimulate mRNA expression of c-fos, insulin-like growth factor-1 and
osteoblastic phenotypes (type I collagen and osteocalcin). Biochem
Biophys Res Commun 199:876880[CrossRef][Medline]
-
Onishi T, Hruska K 1997 Expression of p27Kip1 in
osteoblast-like cells during differentiation with parathyroid hormone.
Endocrinology 138:19950120043
-
Polyak K, Waldman T, He T-C, Kinzler KW, Vogelstein B 1996 Genetic determinants of p53-induced apoptosis and growth arrest. Genes
Dev 10:19451952[Abstract]
-
Santra M, Mann DM, Mercer EW, Skorski T, Calabretta B, Iozzo,
RV 1997 Ectopic expression of decorin protein core causes a generalized
growth suppression in neoplastic cells of various histogenetic origin
and requires endogenous p21, an inhibitor of cyclin-dependent kinases.
J Clin Invest 100:149157[Abstract/Free Full Text]
-
Chen Y, Shyu J-F, Santhanagopal A, Inoue D, David J-P, Dixone
SJ, Horne WC, Baron R, 1998 The calcitonin receptor stimulates Shc
tyrosine phosphorylation and Erk1/2 activation. J Biol Chem 273:1980919816[Abstract/Free Full Text]
-
Houssami S, Findlay DM, Brady CL, Martin TJ, Epand RM, Moore
EE, Murayama E, Tamura T, Orlowski RL, Sexton PM 1995 Divergent
structural requirements exist for calcitonin receptor binding
specificity and adenylate cyclase activation. Mol Pharmacol 47:798809[Abstract]
-
Chen W-J, Armour S, Way J, Chen G, Watson C, Irving P,
Cobb J, Kadwell S, Beaumont K, Rimele T, Kenakin T 1997 Expression
cloning and receptor pharmacology of human calcitonin receptors from
MCF-7 cells and their relationship to amylin receptors. Mol Pharmacol 52:11641175[Abstract/Free Full Text]
-
Nurse P 1990 Universal control mechanism regulating onset of M
phase. Nature 344:503508[CrossRef][Medline]
-
Guadagno TM, Newport JW 1996 Cdk2 kinase is required for entry
into mitosis as a positive regulator of Cdc2-cyclin B kinase activity.
Cell 84:7382[Medline]
-
Zhu L-J, Bagchi MK, Bagchi IC 1998 Attenuation of calcitonin
gene expression in pregnant rat uterus leads to a block in embryonic
implantation. Endocrinology 139:330339[Abstract/Free Full Text]
-
Verheijen MG, Defize LHK 1995 Parathyroid hormone inhibits
mitogen-activation in osteosarcoma cells via a protein kinase
A-dependent pathway. Endocrinology 136:33313337[Abstract]
-
Lee K, Lanske B, Karaplis AC, Deeds JD, Kohno H, Nissenson RA,
Kronenberg HM, Segre GV 1996 Parathyroid hormone-related peptide delays
terminal differentiation of chondrocytes during endochondral bone
development. Endocrinology 137:51095118[Abstract]
-
Lanske B, Karaplis AC, Lee K, Luz A, Vortkamp A, Pirro A,
Karperien M, Defize LHK, Ho C, Mulligan RC, Abou-Samra A-B, Juppner H,
Segre GV, Kronenberg HM 1996 PTH/PTHrP receptor in early development
and Indian hedgehog-regulated bone growth. Science 273:663666[Abstract]
-
Van Biesen T, Luttrell LM, Hawes BE, Lefkowitz RJ 1996 Mitogenic signalling via G protein-coupled receptors. Endocr Rev 17:698714[Medline]
-
Pumiglia MK, Decker SJ 1997 Cell cycle arrest mediated by the
MEK/mitogen-activated protein kinase pathway. Proc Natl Acad Sci USA 94:448452[Abstract/Free Full Text]
-
LaBaer J, Garrett MD, Stevenson LF, Slingerland JM, Sandhu C,
Chou HS, Fattaey A, Harlow E 1997 New functional activities for the p21
family of CDK inhibitors. Genes Dev 11:847862[Abstract]
-
Xiong Y, Hannon GJ, Zhang H, Casso D, Kobayashi R, Beach D 1993 p21 is a universal inhibitor of cyclin kinases. Nature 366:701704[CrossRef][Medline]
-
Li Y, Jenkins CW, Nichols MA, Xiong Y 1994 Cell cycle
expression and p53 regulation of the cyclin-dependent kinase inhibitor
p21. Oncogene 9:22612268[Medline]
-
Pines J 1993 Cyclins and cyclin-dependent kinases: take your
partners. Trends Biochem Sci 18:195197[CrossRef][Medline]
-
Norbury C, Nurse P 1992 Animal cell cycles and their
control. Annu Rev Biochem 61:441470[CrossRef][Medline]
-
Nakano K, Mizuno T, Sowa Y, Orita T, Yoshino T, Okuyama Y,
Fujita T, Ohtani-Fujita N, Matsukawa Y, Tokino T, Yamagishi H, Oka T,
Nomura H, Sakai T 1997 Butyrate activates the WAF1/Cip1 gene promoter
through Sp1 sites in a p53-negative human colon cancer cell line.
J Biol Chem 272:2219922206[Abstract/Free Full Text]
-
Wu R-C, Schonthal AH 1997 Activation of
p53p21waf1 pathway in response to disruption of
cell-matrix interactions. J Biol Chem 272:2909129098[Abstract/Free Full Text]
-
Sheikh MS, Li X-S, Chen JC, Shao ZM, Ordonez JV, Fontana JA 1994 Mechanisms of regulation of WAF1/Cip1 gene expression in human
breast carcinoma: role of p53-dependent and independent signal
transduction pathways. Oncogene 9:34073415[Medline]
-
Rowan S, Ludwig RL, Haupt Y, Bates S, Lu X, Oren M, Vousden KH 1996 Specific loss of apoptotic but not cell-cycle arrest function in a
human tumor derived p53 mutant. EMBO J 15:827838[Abstract]
-
Burgess AMC 1985 The effect of calcitonin on the prechordal
mesoderm, neural plate and neural crest of Xenopus embryo. J
Anat 140:4955[Medline]
-
Wigler M, Silverstein S, Lee LS, Pellicer A, Cheng Y, Axel R 1977 Transfer of purified herpes virus thymidine kinase gene to
cultured mouse cells. Cell 11:223232[Medline]