ArticlePDF Available

The cyclin D1 gene is a target of the β-catenin/LEF-1 pathway

Authors:

Abstract and Figures

beta-Catenin plays a dual role in the cell: one in linking the cytoplasmic side of cadherin-mediated cell-cell contacts to the actin cytoskeleton and an additional role in signaling that involves transactivation in complex with transcription factors of the lymphoid enhancing factor (LEF-1) family. Elevated beta-catenin levels in colorectal cancer caused by mutations in beta-catenin or by the adenomatous polyposis coli molecule, which regulates beta-catenin degradation, result in the binding of beta-catenin to LEF-1 and increased transcriptional activation of mostly unknown target genes. Here, we show that the cyclin D1 gene is a direct target for transactivation by the beta-catenin/LEF-1 pathway through a LEF-1 binding site in the cyclin D1 promoter. Inhibitors of beta-catenin activation, wild-type adenomatous polyposis coli, axin, and the cytoplasmic tail of cadherin suppressed cyclin D1 promoter activity in colon cancer cells. Cyclin D1 protein levels were induced by beta-catenin overexpression and reduced in cells overexpressing the cadherin cytoplasmic domain. Increased beta-catenin levels may thus promote neoplastic conversion by triggering cyclin D1 gene expression and, consequently, uncontrolled progression into the cell cycle.
Content may be subject to copyright.
Proc. Natl. Acad. Sci . USA
Vol. 96, pp. 5522–5527, May 1999
Cell Biology
The cyclin D1 gene is a target of the
b
-cateninyLEF-1 pathway
MICHAEL SHTUTMAN*, JACOB ZHURINSKY*, INBAL SIMCHA*, CHRIS ALBANESE,MARK D’AMICO,
RICHARD PESTELL,AND AVRI BEN-ZEEV*
*Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel; and Department of Medicine and Developmental and
Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
Communicated by Elizabeth D. Hay, Harvard Medical School, Boston, M A, March 15, 1999 (received for review December 30, 1998)
ABSTR ACT
b
-Catenin plays a dual role in the cell: one in
linking the cytoplasmic side of cadherin-mediated cell–cell
contacts to the actin cytoskeleton and an additional role in
signaling that involves transactivation in complex with tran-
scription factors of the lymphoid enhancing factor (LEF-1)
family. Elevated
b
-catenin levels in colorectal cancer caused
by mutations in
b
-catenin or by the adenomatous polyposis
coli molecule, which regulates
b
-catenin degradation, result in
the binding of
b
-catenin to LEF-1 and increased transcrip-
tional activation of mostly unknown target genes. Here, we
show that the cyclin D1 gene is a direct target for transacti-
vation by the
b
-cateninyLEF-1 pathway through a LEF-1
binding site in the cyclin D1 promoter. Inhibitors of
b
-catenin
activation, wild-type adenomatous polyposis coli, axin, and
the cytoplasmic tail of cadherin suppressed cyclin D1 pro-
moter activity in colon cancer cells. Cyclin D1 protein levels
were induced by
b
-catenin overexpression and reduced in cells
overexpressing the cadherin cytoplasmic domain. Increased
b
-catenin levels may thus promote neoplastic conversion by
triggering cyclin D1 gene expression and, consequently, un-
controlled progression into the cell cycle.
b
-Catenin is a major component of adherens junctions linking
the actin cytoskeleton to members of the cadherin family of
transmembrane cell–cell adhesion receptors (1, 2). In addition,
b
-catenin can translocate into the nucleus (3–7), where it can
complex with transcription factors of the LEF-1 family and
regulate the expression of specific genes (8, 9). By playing such
a dual role—a structural role in cell–cell junctions and a
regulatory role in the nucleus—
b
-catenin can transduce
changes in cell adhesion and junction formation to control
transmembrane signaling and gene expression (1, 10–12).
b
-Catenin-mediated signaling depends on its accumulation
and subsequent translocation into the nucleus. The level of
b
-catenin in the cell is regulated by its association with the
tumor suppressor molecule adenomatous polyposis coli (APC;
refs. 13 and 14), axin (15, 16), and glycogen synthase kinase 3
b
(GSK-3
b
; ref. 17). Phosphorylation of
b
-catenin by the APC–
axin–GSK-3
b
complex (18, 19) leads to its degradation by the
ubiquitin–proteasome system (6, 20). The failure of this deg-
radation in cells expressing mutant APC or
b
-catenin leads to
the accumulation of
b
-catenin and is common in human colon
cancer and melanoma (21–23). Elevated
b
-catenin levels in
such tumors are suggested to confer uncontrolled activation of
gene transcription by the
b
-cateninyLEF-1 complex that may
contribute to tumor progression (1, 24, 25). The nature of the
target genes of the
b
-cateninyLEF complex is, however, largely
unknown, except for the recently discovered c-MYC that was
shown to contain LEF-1-binding sequences in its promoter (26).
Cyclin D1 is a major regulator of the progression of cells into
the proliferative stage of the cell cycle (27). Although the
cyclin D1 gene is not amplified in human colon cancer, the
expression of cyclin D1 is elevated in about 30% of human
adenocarcinomas and in adenomatous polyps of the colon (28,
29), and expression of anti-sense cyclin D1 cDNA abolished
the growth of SW480 colon cancer cells in nude mice, indi-
cating a critical role for cyclin D1 in tumorigenesis (30).
In this study, we investigated the possibility that the cyclin
D1 gene is a target for the
b
-cateninyLEF-1 complex and show
that the cyclin D1 promoter contains a LEF-1 binding se-
quence that is activated in human colon cancer cells. We
further show that transcriptional activation of the cyclin D1
gene in such cells can be inhibited by enhancing the degrada-
tion of
b
-catenin with wild-type APC and axin or by its binding
to the cadherin cytoplasmic tail. Unscheduled activation of
cyclin D1 transcription by the
b
-cateninyLEF-1 complex in
colon cancer cells may result in uncontrolled cell proliferation
and thus contribute to tumor progression in these cells.
MATERIALS AND METHODS
Plasmid Constructions. Plasmids containing the luciferase
reporter under the cyclin D1 promoter and the deletion
constructions derived from it have been described (31). The
mutated LEF-1 binding site at nucleotides 275 and 274 from
AT to GC (2163mtLefCD1LUC) and the deletion of the
LEF-1 binding site from 281 to 273 (2163DLefCD1LUC)
were constructed by PCR-based site directed mutagenesis by
using the 2163CD1LUC plasmid. Reporter plasmids contain-
ing luciferase under a multimeric consensus- or mutant inac-
tive-LEF-1 binding site (TOPFLASH and FOPFLASH; ref. 9)
and vectors expressing various forms of
b
-catenin (7), LEF-1
(5), axin, APC (15), and the N-cadherin tail (32) have been
described.
Cell Lines and Transfections. SW480, 293T, and Neuro 2A
cell lines were maintained in DMEM with 10% (volyvol) calf
serum. Transient transfections were performed by the calcium
phosphate precipitation method with 293T and Neuro 2A cell
lines and by Lipofectamine with SW480 cells. A
b
-galactosi-
dase-expressing plasmid (0.5
m
g) was included in each trans-
fection to monitor the transfection efficiency. We found that
b
-galactosidase expression was not affected significantly by
either LEF-1 or
b
-catenin cotransfection. After 48 h, the cells
were lysed, and luciferase and
b
-galactosidase activities were
determined by enzyme assay kits from Promega. Luciferase
activity was normalized to
b
-galactosidase activity as an in-
ternal transfection control. N-cadherin-tail-expressing cell
lines were generated by stably transfecting the pECE-N-
cadherin tail and pSV-hygro expression vectors into SW480
cells (7, 32). Individual clones were selected for resistance to
100
m
gyml hygromycin. The green fluorescent protein (GFP)–
DN-
b
-catenin and the mutant
b
-catenin constructs (
b
-cat Y33)
were HA-tagged (7).
The publication costs of this article were defrayed in part by page charge
payment. This article must therefore be hereby marked ‘‘advertisement’’ in
accordance with 18 U.S.C. §1734 solely to indicate this fact.
PNAS is available online at www.pnas.org.
Abbreviations: APC, adenomatous polyposis coli; GSK-3
b
, glycogen
synthase kinase 3
b
; GFP, green fluorescent protein.
To whom reprint requests should be addressed. e-mail: lgbenzev@
weizmann.weizmann.ac.il.
5522
Immunoblotting. Protein levels were determined by immu-
noblotting by using a monoclonal antibody against cyclin D1
(DCS-6; Neomarkers, Freemont, CA), a polyclonal antiserum
against
b
-catenin, and monoclonal antibodies against pan
cadherin (CH-19), and vinculin (h-VIN 1) as described (ref. 7;
all from Sigma). Anti-LEF-1 rabbit antiserum was a gift from
R. Grosschedl (University of California, San Francisco).
In Vitro Translation and DNA Binding Analysis. In vitro
translated proteins were prepared by using a coupled transcrip-
tion and translation kit (Promega).
b
-Catenin and LEF-1 were
expressed from pCIneo constructs, and the efficiency of trans-
lation was determined by a reaction containing [
35
S]methionine,
followed by SDSyPAGE and Western blotting. For protein–DNA
interaction, in vitro translated LEF-1 and
b
-catenin were incu-
bated with the following
32
P-labeled duplex oligonucleotide
probes: CD1 (59-CTCTGCCGGGCTTTGATCTTTGCTTAA-
CAACA-39), CD1TOP (59-CTCTGCCGGCCTTTGATCTTT-
GCTTAACAACA-39), and CD1FOP (59-CTCTGCCGGGCT-
TTGGCCTTTGCTTAACAACA-39). The wild-type and mu-
tant LEF-1 binding sequences are underlined. The binding
reaction contained '40,000 cpm of
32
P-labeled DNA that was
incubated for 30 min with the in vitro translated proteins in 20 mM
Hepes, pH 7.9y75 mM NaCly1 mM dithiotreitoly2 mM MgCl
2
y
10% (vol/vol) glyceroly0.1 mg of BSAy10
m
g/ml salmon sperm
DNA. DNA–protein complexes were electrophoresed in 4%
native acrylamide gels and visualized by autoradiography. Bind-
ing reactions also were carried out with nuclear extracts from
SW480 cells (3
m
g protein per reaction mixture) that were
prepared as described (33).
RESULTS
Activation of the Cyclin D1 Promoter and Elevation of
Cyclin D1 in Cells Transfected with
b
-Catenin. We assessed
the effect of
b
-catenin overexpression on cyclin D1 abundance
in human 293T cells, previously shown to respond by activation
of
b
-cateninyLEF-1-driven transcription (7, 21, 22). Cells were
transfected with expression vectors encoding mutant N-
terminal
b
-catenin constructs (GFP–DN-
b
-catenin and
b
-cat
Y33) that render it stable against degradation (7, 22, 23).
Cyclin D1 protein levels were induced 2-to 3-fold after trans-
fection with these
b
-catenin constructs, as compared with
transfection with a construct encoding the GFP that served as
control (Fig. 1A). To examine whether the cyclin D1 promoter
was a direct transcriptional target for activation by the
b
-cate-
ninyLEF-1 complex, an HA-tagged DN-
b
-catenin expression
plasmid (pHA-DN-
b
-catenin) was cotransfected with the hu-
man cyclin D1 promoter linked upstream to a luciferase
reporter (Fig. 2A). The cyclin D1 promoter was induced
12-fold by this
b
-catenin (Fig. 1B) and was not affected by
either the empty expression plasmid (Fig. 1B; pA3) or the
construct encoding GFP. The cyclin D1 promoter was also
induced 10- to 15-fold by
b
-catenin overexpression in the
human Neuro 2A neuroblastoma cell line (see below; Fig. 3).
Identification of a LEF-1 Binding Sequence in the Cyclin D1
Promoter. To identify the sequences in the cyclin D1 promoter
that can confer transactivation by
b
-catenin, a series of 59
promoter deletion constructions were used (Fig. 2 A). Such
mapping identified a region between nucleotides 2141 and
266 that was sufficient for transcriptional activation by
b
-cate-
nin (Fig. 2B). Analysis of the sequence between nucleotides
2141 and 266 indicated that nucleotides 281 to 273 repre-
sent a consensus LEF binding site (Fig. 2 A,Insert). To examine
the functional significance of this candidate LEF binding site,
we introduced (in the context of the 2163 bp reporter plasmid,
2163CD1LUC) a deletion (2163DLefCD1LUC) and point
mutations (163mtLefCD1LUC) that should render the LEF
binding site in the cyclin D1 promoter inactive (Fig. 3B).
b
-Catenin-driven transactivation of the cyclin D1 constructs
was inhibited when this putative LEF-1-binding domain was
either deleted or mutated (Fig. 3A). Mutations introduced
outside of this region and in the Sp1 binding site of this
construct (2163DSp1CD1LUC; ref. 34) had no effect on
b
-catenin-driven activation of the promoter (Fig. 3A). Trans-
activation of the cyclin D1 promoter by
b
-catenin also de-
pended on the level of cotransfected LEF-1. At low concen-
trations of the expression plasmid, LEF-1 elevated
b
-catenin-
mediated activation of cyclin D1 a further 2.5-fold (Fig. 3C) but
inhibited transcription at high plasmid concentrations, consis-
tent with its transcription repressor role when expressed in
excess over
b
-catenin (35, 36).
The
b
-CateninyLEF-1 Complex Binds in Vitro to the LEF-1
Consensus Sequence of the Cyclin D1 Promoter. To determine
whether the LEF-1 binding sequence in the cyclin D1 promoter
can bind a LEF-1y
b
-catenin heterodimer, we conducted elec-
trophoretic mobility-shift assays comparing the cyclin D1
LEF-1 site with a consensus LEF-1-binding site (CD1TOP; ref.
9) and a mutant LEF-1 binding site (CD1FOP). In vitro,
synthesized LEF-1 bound to the LEF-1 sequence in the cyclin
D1 promoter (Fig. 4A, CD1, lane 2), and the complex was
FIG. 1. Induction of cyclin D1 and
b
-catenin-responsive transac-
tivation of the cyclin D1 promoter. (A) Cells from the 293T line were
transfected with 4
m
g of GFP, GFP-linked DN-
b
-catenin (GFP-DN-
b
-cat), or mutant
b
-cat Y33. Cell lysates were analyzed by Western
blotting for levels of cyclin D1,
b
-catenin (
b
-cat), and vinculin. (B)
Cells from the 293T line were transfected with 0.8
m
g of a reporter
plasmid containing 1,745 bp of the cyclin D1 promoter
(21745CD1Luc) or with the empty pA3 plasmid, together with 4
m
g
of an HA-tagged DN-
b
-catenin-encoding plasmid (pHA-DN-
b
-cat), a
GFP expression construct, or pCIneo. The bars represent luciferase
activity in cells transfected with pHA-DN-
b
-catenin divided by the
activity in cells transfected with control plasmid (pCIneo). Each
transfection was carried out in duplicate plates. The means 6SD from
three separate transfections are shown.
Cell Biology: Shtutman et al.Proc. Natl. Acad. Sci. USA 96 (1999) 5523
supershifted with an LEF-1 antibody (Fig. 4 A, lanes 4 and 8).
The mutant sequence derived from the 2163mtLefCD1LUC
(CD1FOP), in contrast, failed to bind LEF-1 (Fig. 4A, CD1FOP,
lane 10).
b
-Catenin transcribed and translated in vitro did not bind
on its own, as expected, to this sequence in the cyclin D1 promoter
(Fig. 4A, lanes 1, 5, and 9). A supershift was introduced in the
electrophoretic mobility of this DNA fragment when
b
-catenin
and LEF-1 were added together (Fig. 4A, lanes 3 and 7),
implying the formation of a ternary complex consisting of the
DNA, LEF-1, and
b
-catenin. This binding of LEF-1 to the
cyclin D1 promoter was blocked by the addition of excess
unlabeled CD1 but not by the mutant CD1FOP (Fig. 4B).
Transactivation of the Cyclin D1 Promoter in Colon Cancer
Cells and Its Inhibition by APC, Axin, and the Cadherin
Cytoplasmic Domain. To analyze effects on transcription of
the cyclin D1 promoter in a cellular system where endogenous
b
-catenin is elevated, we employed SW480 colon carcinoma
cells in which the level of
b
-catenin is increased because of
inactivating mutations in APC (37). Nuclear extracts from
these cells could specifically bind to and shift the electro-
phoretic mobility of the LEF-1 binding sequence in the cyclin
D1 promoter (Fig. 5A, lane 3), similar to in vitro translated
LEF-1 (Fig. 5A, lane 1). An additional shifted band (Fig. 5A,
marked X) bound nonspecifically; it also was observed when
the mutant LEF-1 binding site (CD1FOP) was used (Fig. 5A,
lanes 6– 8).
When the cyclin D1 promoter, in the context of the
2163CD1LUC reporter plasmid, was transfected into SW480
cells, transcription from this plasmid was active without co-
transfection of exogenous
b
-catenin (Fig. 5B). This transcrip-
tion was reduced by cotransfection of wild-type APC or a
mixture of APC and axin (Fig. 5B), which act together to
decrease levels of
b
-catenin by enhancing its degradation (15,
16). In contrast, APC and axin did not affect the basal
transcription observed in the deletion mutant of this cyclin D1
promoter construct (2163DLefCD1LUC; Fig. 5B). We have
FIG. 2. Identification of the LEF-1 binding sequence in the cyclin
D1 promoter. (A) Schematic representation of reporter constructs
from the cyclin D1 promoter, deletion constructs, and the LEF-1-
binding sequence between nucleotides 281 and 273 of the promoter
(Insert). (B) The promoter deletion constructs of the cyclin D1
promoter (0.8
m
g) shown in Awere transfected into 293T cells as
described in Fig. 1B.
FIG. 3. The LEF-1-binding sequence in the cyclin D1 promoter is
required for
b
-catenin-mediated transactivation. (A) Cells from the
293T line were transfected with the indicated reporter plasmid (0.8
m
g), together with 4
m
gofDN-
b
-catenin or the control plasmid
(pCIneo). The promoter activity is presented as in Fig. 1B.(B)
Schematic representation of mutations in the 2163 cyclin D1 pro-
moter construct, including an AT to GC change at nucleotides 275 and
274 (2163mtLefCD1LUC) and deletion of the LEF-1 binding site
(2163DLefCD1LUC) between nucleotides 281 to 273. (C) Neuro 2A
cells were transfected with 0.8
m
gof21745CD1Luc, 2
m
gofDN-
b
-
catenin, or a control plasmid (pCIneo), along with increasing amounts
of a LEF-1 expression plasmid. DNA concentrations were kept
constant with empty vector DNA. Transfections were carried out in
triplicate and the means 6SD are presented.
5524 Cell Biology: Shtutman et al.Proc. Natl. Acad. Sci. USA 96 (1999)
shown previously that either full-length N-cadherin or its
cytoplasmic domain can block the constitutive
b
-catenin-
mediated transactivation in SW480 cells by competing with
LEF-1 for
b
-catenin binding (7, 32). We therefore analyzed the
effect of a stably overexpressed N-cadherin cytoplasmic tail in
SW480 cells on the transcriptional activity of the cyclin D1
promoter and the level of the cyclin D1 protein. In SW480 cells
stably expressing high levels of the cadherin cytoplasmic tail
(Fig. 6B, clone A8) but not in clones expressing low levels of
the cadherin tail (Fig. 6B, clone A20),
b
-catenin-mediated
transactivation of the cyclin D1 promoter was inhibited (Fig.
6A; compare clones A8 and A20), and the level of cyclin D1
protein was reduced specifically (Fig. 6B; compare clones A8
and A20).
FIG. 4. Electrophoretic mobility-shift assays of the cyclin D1
promoter. (A) Duplex oligonucleotides containing the LEF-1 binding
sequences of the cyclin D1 promoter (CD1), a consensus LEF-1
binding sequences (CD1TOP) and a substitution of nucleotides 275
and 274 from AT to GC (CD1FOP) were ‘‘end labeled’’ with
[
32
P]dATP and incubated with in vitro translated LEF-1 andyor
b
-catenin or with anti-LEF-1 antibody. The protein–DNA complexes
were separated by electrophoresis and visualized by autoradiography.
(B)In vitro translated LEF-1 was incubated with the
32
P-labeled CD1
oligonucleotide, and increasing amounts of unlabeled CD1 or
CD1FOP oligonucleotides were used as competitors. The protein–
DNA complexes were analyzed as in Fig. 4A.
FIG. 5. Electrophoretic mobility-shift assays of the cyclin D1
promoter with SW480 nuclear lysates and regulation of cyclin D1
promoter transcriptional activity by APC and axin. (A) Oligonucleo-
tides labeled with [
32
P]dATP (CD1, lanes 1–5; CD1FOP, lanes 6–8)
were incubated with in vitro translated LEF-1 (lanes 1 and 2) or with
nuclear extracts from SW480 cells (lanes 3–8) in the presence of
100-fold excess of cold competitor oligonucleotides. (B) SW480 cells
were transiently transfected with 1
m
gof2163CD1LUC or
2163DLefCD1LUC and 2
m
g of either GFP or APC plus axin
expression vectors. The bars represent luciferase activity in the trans-
fected cells after normalizing for transfection efficiency with
b
-galac-
tosidase. Note that, in A, an additional, faster-migrating band was
obtained with CD1 and the SW480 nuclear extract (X). This band also
was seen with the mutant CD1FOP and therefore was considered
nonspecific.
Cell Biology: Shtutman et al.Proc. Natl. Acad. Sci. USA 96 (1999) 5525
DISCUSSION
Elevated
b
-catenin expression in colorectal cancer caused by
inactivating mutations in APC or in the GSK-3
b
phosphory-
lation sites of
b
-catenin results in the accumulation of
b
-cate-
nin in the nucleus and the uncontrolled activation of target
gene expression—a process believed to contribute to tumor
progression. The nature of these target genes is still largely
unknown, but their involvement in the control of cell prolif-
eration is suggested. In this study, we have identified the cyclin
D1 gene as a target for the
b
-cateninyLEF-1 complex. We have
shown that the LEF-1 binding consensus sequence in the cyclin
D1 gene promoter can bind to nuclear extracts from human
colon cancer cells and is transcriptionally active when linked to
reporter plasmids and introduced into human colon cancer
cells expressing elevated wild-type
b
-catenin. In such cells
expressing inactive mutant APC, cotransfection with wild-type
APC inhibited transactivation of the cyclin D1 gene. Based on
these results, we suggest that increased
b
-catenin levels in
colon cancer cells result in the activation of the cyclin D1 gene
promoter by the heterodimeric complex formed between
b
-catenin and LEF-1, which, in turn, results in the elevation of
cyclin D1 gene expression and protein level. This elevation, in
turn, may lead to uncontrolled stimulation of the cells into the
proliferative stage of the cell cycle. In the presence of wild-t ype
APC, the level of
b
-catenin is kept in check, and the formation
of the
b
-cateninyLEF-1 complex is prevented. The reduction
in the transcriptional activity of the cyclin D1 promoter,
resulting most probably from competition between the cyto-
plasmic tail of cadherin and LEF-1 for
b
-catenin binding (32),
and the concomitant decrease in cyclin D1 protein support this
view (Figs. 5 and 6). These results also show the potential
usefulness of the cadherin cytoplasmic domain (and its deriv-
atives) in the attempt to antagonize the possibly oncogenic
transcription driven by the
b
-cateninyLEF-1 complex (32).
Elevated transcription from the cyclin D1 gene induced by
the
b
-cateninyLEF-1 complex may also explain the notion
that, although alterations in the cyclin D1 gene were detected
in many types of human cancer (3840), no mutations in the
cyclin D1 gene were identified in colorectal cancer, whereas
'30% of primary human colon tumors display increased
expression of cyclin D1 (28, 29). This increase in cyclin D1 in
only some colon cancers may result from the heterogeneity in
b
-catenin expression in the tumor and from the fact that many
colon cancer cell lines with mutant APC do not display
increased levels of
b
-catenin (41).
The involvement of cyclin D1 in human colon cancer also is
supported by experiments showing that expression of an
antisense cyclin D1 construct in SW480 colon cancer cells (also
used in this study) results in decreased cyclin D1, inhibition of
growth, and suppression of their tumorigenicity in nude
mice (30).
b
-Catenin and cyclin D1 apparently also share another
component of the Wnt pathway, GSK-3
b
. A recent study
showed that cyclin D1 is a target for GSK-3
b
, which, by
regulating cyclin D1 phosphorylation, may control the subcel-
lular localization and proteolysis of cyclin D1 by the ubiquitin–
proteasome system (42). This pathway may constitute another
means by which the Wnt signaling pathway can target cyclin D1
in colon cancer.
The link between
b
-catenin-mediated transactivation of the
cyclin D1 promoter and neoplastic transformation also may be
relevant to intestinal and mammary epithelial cells trans-
formed by the elevated expression of integrin-linked kinase
(ILK) (43). ILK-transfected cells were shown to become
neoplastic and to display high levels of cyclin D1 protein
constitutively (44). In addition, the level of extrajunctional
b
-catenin in such cells is increased, concomitant with nuclear
translocation of
b
-catenin and activation of
b
-catenin-
mediated LEF-1-directed transcription (45). Cyclin D1 eleva-
tion in these ILK-transfected cells may result from a direct
activation of cyclin D1 transcription by the
b
-cateninyLEF-1
complex.
In a recent study, the c-MYC promoter was identified also as
a target of the
b
-cateninyLEF-1 complex in human colorectal
cancer (26). In other types of tumors, overexpression of either
c-myc or cyclin D1 were shown to induce cell-cycle progression
in breast cancer cell lines (46), whereas, in fibroblasts, myc
induction of cell-cycle progression seems to be linked to the
induction of cyclin E, independently of cyclin D1 (47). To-
gether, these results are consistent with a model in which myc
and cyclin D1 can induce S phase entry in parallel pathways,
and the elevation of cyclin D1 in colorectal cancer, by inacti-
vation of APC or mutations in
b
-catenin, may promote
uncontrolled proliferation and thus contribute to the neoplas-
tic transformation of cells.
We are grateful to the following colleagues for sending reagents: S.
Sokol, R. Kemler, H. Clevers, M. van de Wetering, M. Wheelock, and
R. Grosschedl. These studies were supported by grants from the
USA–Israel Binational Foundation, the German–Israeli Foundation
for Scientific Research and Development, the Forchheimer Center for
Molecular Genetics, the Cooperation Program in Cancer Research
between the German Cancer Research Center and the Israel Ministry
of Science (to A.B.-Z.), and from the National Institute of Health (to
R.G.P.). A.B.-Z. holds the Lunenfeld–Kunin Chair in Cell Biology and
Genetics, and R.G.P. is a recipient of the Ira T. Hirschl Award and the
Susan G. Komen Breast Cancer Foundation Award.
1. Ben-Ze’ev, A. & Geiger, B. (1998) Curr. Opin. Cell Biol. 10,
629– 639.
2. Kemler, R. (1993) Trends Genet. 9, 317–321.
FIG. 6. Decreased transcription from the cyclin D1 promoter and
cyclin D1 protein in SW480 cells expressing the N-cadherin cytoplas-
mic domain. (A) Individual SW480 cell clones stably expressing
different levels of the N-cadherin cytoplasmic domain were transiently
transfected with 1
m
gof21745CD1LUC. The bars represent luciferase
activity in cells transfected with 21745CD1LUC divided by luciferase
activity in cells transfected with the empty pCIneo vector. (B) Total
cellular proteins from the cells described in Awere separated by
SDSyPAGE and analyzed by Western blotting with antibodies that
recognize the N-cadherin tail,
b
-catenin, and cyclin D1.
5526 Cell Biology: Shtutman et al.Proc. Natl. Acad. Sci. USA 96 (1999)
3. Behrens, J., von Kries, J. P., Kuhl, M., Bruhn, L., Wedlich, D.,
Grosschedl, R. & Birchmeier, W. (1996) Nature (London) 382,
638– 642.
4. Molenaar, M., van de Wetering, M., Oosterwegel, M., Peterson-
Maduro, J., Godsave, S., Korinek, V., Roose, J., Destree, O. &
Clevers, H. (1996) Cell 86, 391–399.
5. Huber, O., Korn, R., McLaughlin, J., Ohsugi, M., Herrmann,
B. G. & Kemler, R. (1996) Mech. Dev. 59, 3–11.
6. Salomon, D., Sacco, P. A., Guha Roy, S., Simcha, I., Johnson,
K. R., Wheelock, M. J. & Ben-Ze’ev, A. (1997) J. Cell Biol. 139,
1325–1335.
7. Simcha, I., Shtutman, M., Salomon, D., Zhurinsky, J., Sadot, E.,
Geiger, B. & Ben-Ze’ev, A. (1998) J. Cell Biol. 141, 1433–1448.
8. Riese, J., Yu, X., Munnerlyn, A., Eresh, S., Hsu, S.-C., Gros-
schedl, R. & Bienz, M. (1997) Cell 88, 777–787.
9. van de Wetering, M., Cavallo, R., Dooijes, D., van Beest, M., van
Es, J., Loureiro, J., Tpma, A., Hursh, D., Jones, T., Bejsovec, A.,
Peifer, M., Mortin, M. & Clevers, H. (1997) Cell 88, 789–799.
10. Cavallo, R., Rubenstein, D. & Peifer, M. (1997) Curr. Opin.
Genet. Dev. 7, 459- 466.
11. Bullions, L. C. & Levine, A. (1998) Curr. Opin. Oncol. 10, 81–87.
12. Willert, K. & Nusse, R. (1998) Curr. Opin. Genet. Dev. 8, 95–102.
13. Su, L. K., Vogelstein, B. & Kinzler, K. W. (1993) Science 262,
1734–1737.
14. Rubinfeld, B., Souza, B., Albert, I., Mu¨ller, O., Chamberlain,
S. F., Masiarz, R., Munemitsu, S. & Polakis, P. (1993) Science 262,
1731–1734.
15. Hart, M., de los Santos, R., Albert, I., Rubinfeld, B. & Polakis,
P. (1998) Curr. Biol. 8, 573–581.
16. Behrens, J., Jerchow, B. A., Wurtele, M., Grimm, J., Asbrand, C.,
Wirtz, R., Kuhl, M., Wedlich, D. & Birchmeier, W. (1998) Science
280, 596–599.
17. Rubinfeld, B., Albert, I., Porfiri, E., Fiol, C., Munemitsu, S. &
Polakis, P. (1996) Science 272, 1023–1026.
18. Yost, C., Torres, M., Miller, J. R., Huang, E., Kimelman, D. &
Moon, R. T. (1996) Genes Dev. 10, 1443–1454.
19. Ikeda, S., Kishida, S., Yamamoto, H., Murai, H., Koyama, S. &
Kikuchi, A. (1998) EMBO J. 17, 1372–1384.
20. Aberle, H., Bauer, A., Stappert, J., Kispert, A. & Kemler, R.
(1997) EMBO J. 16, 3797–3804.
21. Morin, P. J., Sparks, A. B., Korinek, V., Barker, N., Clevers, H.,
Vogelstein, B. & Kinzler, K. W. (1997) Science 275, 1787–1790.
22. Korinek, V., Backer, N. P., Morin, J., van Wichen, D., de Weger,
R., Kinzler, K. W., Vogelstein, B. & Clevers, H. (1997) Science
275, 1784–1787.
23. Rubinfeld, B., Robbins, P., El-Gamil, M., Albert, I., Porfiri, E. &
Polakis, P. (1997) Science 275, 1790–1792.
24. Peifer, M. (1997) Science 275, 1752–1753.
25. Gumbiner, B. M. (1997) Curr. Biol. 7, R443–R446.
26. He, T. C., Sparks, A. B., Rago, C., Hermeking, H., Zawel, L., da
Costa, L. T., Morin, P. J., Vogelstein, B. & Kinzler, K. W. (1998)
Science 281, 1509–1512.
27. Sherr, C. J. (1996) Science 274, 1672–1677.
28. Bartkova, J., Lukas, J., Strauss, M. & Bartek, J. (1994) Int. J.
Cancer 58, 568–573.
29. Arber, N., Hibshoosh, H., Moss, S. F., Sutter, T., Zhang, Y., Begg,
M., Wang, S., Weinstein, I. B. & Holt, P. R. (1996) Gastroenter-
ology 110, 669674.
30. Arber, N., Doki, Y., Han, E. K.-H., Sgambato, A., Zhou, P., Kim,
N.-H., Delohery, T., Klein, M. G., Holt, P. R. & Weinstein, I. B.
(1997) Cancer Res. 57, 1569–1574.
31. Albanese, C., Johnson, J., Watanabe, G., Eklund, N., Vu, D.,
Arnold, A. & Pestell, R. G. (1995) J. Biol. Chem. 270, 23589
23597.
32. Sadot, E., Simcha, I., Shtutman, M., Ben-Ze’ev, A. & Geiger, B.
(1998) Proc. Natl. Acad. Sci . USA 95, 15339–15344.
33. Schreiber, E., Matthias, P., Mu¨ller, M. M. & Schaffner, W. (1989)
Nucleic Acids Res. 17, 6419.
34. Watanabe, G., Albanese, C., Lee, J. R., Reutens, A., Vairo, G.,
Henglein, B. & Pestell, R. G. (1998) Mol. Cell. Biol. 18, 3212–
3222.
35. Cavallo, R. A., Cox, R. T., Moline, M. M., Roose, J., Polevoy,
G. A., Clevers, H., Peifer, M. & Bejsovec, A. (1998) Nature
(London) 395, 604– 608.
36. Roose, J., Molenaar, M., Peterson, J., Hurenkamp, J., Brantjes,
H., Moerer, P., van de Wetering, M., Destree, O. & Clevers, H.
(1998) Nature (London) 395, 607–612.
37. Munemitsu, S., Albert, I., Souza, B., Rubinfeld, B. & Polakis, P.
(1995) Proc. Natl. Acad. Sci . USA 92, 3046–3050.
38. Hunter, T. & Pines, J. (1994) Cell 79, 573–582.
39. Motokura, T. & Arnold, A. (1993) Curr. Opin. Genet. Dev. 3,
5–10.
40. Hall, M. & Peters, G. (1996) Adv. Cancer Res. 68, 67–108.
41. Polakis, P. (1997) Biochim. Biophys. Acta 1332, F127–F147.
42. Diehl, A. J., Cheng, M., Rousell, M. F. & Sherr, C. J. (1998) Genes
Dev. 12, 3499–3511.
43. Hanigan, G. E., Leung-Hagesteijn, C., Fitz-Gibbon, L., Coppo-
lino, M. G., Radeva, G., Filmus, J., Bell, J. C. & Dedhar, S. (1996)
Nature (London) 379, 91–96.
44. Radeva, G., Petrocelli, T., Behrend, E., Leung-Hagelsteijn, C.,
Filmus, J., Slingerland, J. & Dedhar, S. (1997) J. Biol. Chem. 272,
13937–13944.
45. Novak, A., Hsu, S.-C., Leung-Hagelsteijn, C., Radeva, G., Pap-
koff, J., Montesano, R., Roskelley, C., Grosschedl, R. & Dedhar,
S. (1998) Proc. Natl. Acad. Sci . USA 95, 4374– 4379.
46. Prall, O. W., Rogan, E. M., Musgrove, E. A., Watts, C. K. &
Sutherland, R. L. (1998) Mol. Cell. Biol. 18, 4499 4508.
47. Vlach, J., Hennecke, S. & Amati, B. (1996) EMBO J. 15,
6595–6604.
Cell Biology: Shtutman et al.Proc. Natl. Acad. Sci. USA 96 (1999) 5527
... Gene ontology analysis of differentially expressed genes (DEGs) identified enrichment of genes regulated by β-catenin (CTNNB1) when comparing AFs vs. FCs, but not TMs vs. MCs (Fig. 5B, below). Hierarchical clustering of the PCDH19 cohort based on significantly dysregulated Wnt/β-catenin targets [APCDD1 [38], AR [39], CCND1 [40], CYP24A1 [41], EPB41L4A [42], INHBB [43], MMP1 [44] and SERPINA3 [45]] and FZD3 revealed that AFs have Wnt/β-catenin target expression more similar to male than female controls, while TMs cluster among the AFs (Fig. 5B, above). Altogether, these results point towards dysregulation of the Wnt/β-catenin signalling pathway in individuals with pathogenic PCDH19 variants. ...
Article
Full-text available
Clustering Epilepsy (CE) is a neurological disorder caused by pathogenic variants of the Protocadherin 19 (PCDH19) gene. PCDH19 encodes a protein involved in cell adhesion and Estrogen Receptor α mediated-gene regulation. To gain further insights into the molecular role of PCDH19 in the brain, we investigated the PCDH19 interactome in the developing mouse hippocampus and cortex. Combined with a meta-analysis of all reported PCDH19 interacting proteins, our results show that PCDH19 interacts with proteins involved in actin, microtubule, and gene regulation. We report CAPZA1, αN-catenin and, importantly, β-catenin as novel PCDH19 interacting proteins. Furthermore, we show that PCDH19 is a regulator of β-catenin transcriptional activity, and that this pathway is disrupted in CE individuals. Overall, our results support the involvement of PCDH19 in the cytoskeletal network and point to signalling pathways where PCDH19 plays critical roles.
... Moreover, many Wnt response elements were authenticated in the c-MYC promoter (Rennoll and Yochum 2015). Cyclin D1 is another direct target of β-catenin and may be an important molecule that activates β-catenin to promote cell proliferation (Shtutman et al. 1999;Tetsu and McCormick 1999;Delgado et al. 2013). Multiple reports have suggested that activated Wnt/β-catenin triggers Cyclin D1 expression in mouse and human HCC (Delgado et al. 2013;Kaur et al. 2012). ...
Article
Full-text available
The function of Biliverdin Reductase A (BLVRA) in hepatocellular carcinoma (HCC) cells proliferation, invasion and migration remains unclear. Therefore, this research intends to explore the effect of BLVRA on HCC cells growth and metastasis. BLVRA expression was analyzed in public dataset and examined by using western blot. The malignant function of BLVRA in HCC cell lines and its effect on Wnt/β-catenin pathway were measured. Analysis from GEPIA website showed that BLVRA expression was significantly increased in HCC tissues, and high expression of BLVRA resulted in worse prognosis of HCC patients. Results from western blot showed that BLVRA expression was obviously increased in HCC cell lines. Moreover, HepG2 and Hep3B cells in si-BLVRA-1 or si-BLVRA-2 group displayed an obvious reduction in its proliferation, cell cycle, invasion and migration compared to those in the si-control group. Additionally, si-BLVRA-1 or si-BLVRA-2 transfection significantly reduced the protein levels of Vimentin, Snail1 and Snail2, as well as decreased Bcl-2 expression and increased Bax and cleaved-caspase 3 expression. Furthermore, si-BLVRA treatment inhibited the protein levels of c-MYC, β-catenin, and Cyclin D1. After IWP-4 (Wnt/β-catenin inhibitor) treatment, the proliferation ability of HCC cells was significantly reduced. BLVRA expression was significantly increased in HCC tissues and cell lines, and knocked down of BLVRA could suppress the proliferation, invasion and migration in HCC cell lines, as well as induce cell apoptosis. Moreover, si-BLVRA transfection blocked the activation of Wnt/β-catenin pathway.
... Activation of the canonical Wnt/β-catenin signaling involves the formation of a ternary complex composed of Wnt, FZD, and the co-receptor low density lipoprotein receptor-related protein 5 or 6 (LRP5/6). Wnt stimulation results in the stabilization and 4 nuclear import of β-catenin, which acts as a transcriptional co-activator interacting with members of the T cell factor/lymphoid enhancer binding factor (TCF/LEF) family of transcription factors to induce the expression of Wnt target genes (e.g., CyclinD1 and Axin2 (Shtutman et al. 1999;Jho et al. 2002;Lustig et al. 2002)). The Wnt/β-catenin pathway controls cell fate decisions during embryonic development and in adult organisms, acting as a stem cell niche signal in many tissues (Gordon and Nusse 2006;Nusse and Clevers 2017). ...
Preprint
Full-text available
In the dentate gyrus of the adult hippocampus, neurogenesis from neural stem cells (NSCs) is regulated by Wnt signals from the local microenvironment. The Wnt/β-catenin pathway is active in NSCs, where it regulates proliferation and fate commitment, and subsequently its activity is strongly attenuated. The mechanisms controlling this pattern of activity are poorly understood. In stem cells from adult peripheral tissues, secreted R-spondin proteins (RSPO1-4) interact with LGR4-6 receptors and control Wnt signaling strength. Here, we found that RSPO1-3 and LGR4-6 are expressed in the adult dentate gyrus and in cultured NSCs isolated from the adult mouse hippocampus. The expression of LGR4-5 decreased in NSCs upon differentiation, concomitantly with the reported decrease in Wnt activity. Treatment with RSPO1-3 increased hippocampal NSCs proliferation and the expression of the Wnt target gene Cyclin D1. Moreover, RSPO1-3 were expressed by primary cultures of dentate gyrus astrocytes, a crucial component of the neurogenic niche able to induce NSC proliferation and neurogenesis. In co-culture experiments, astrocyte-induced proliferation of NSCs was prevented by RSPO2 knockdown in astrocytes, and by LGR5 knockdown in hippocampal NSCs. Altogether, our results indicate that RSPO/LGR signaling is present in the dentate niche, where it could control Wnt activity and proliferation of NSCs.
... So, we further investigated the expression of some β-catenin target genes and the correlativity between TC1 or Chibby and them. These target genes have been implicated in cancer proliferation, such as c-Myc [27] and Cyclin D1, [28] and in cancer invasion, such as MMP-7. [29] Our Figure 4. Correlations between TC1, Chibby, β-catenin and overall survival rate in LSCC patients. ...
Article
Full-text available
As an important member of Wnt/β-catenin signaling pathway, the aberrant expression of β-catenin has been implicated in many cancers. Chibby, a β-catenin binding partner, is an antagonist involved in this pathway. In contrast, thyroid cancer 1 (TC1) as an activator of this pathway can relieve the antagonistic activity of Chibby on the β-catenin-mediated transcription and is high expressed in human tumors. The objectives of this study were to examine the expression of TC1, Chibby, and β-catenin and investigate the association among them in laryngeal squamous cell carcinoma (LSCC). The expression of TC1, Chibby, β-catenin, c-Myc, Cyclin D1, and matrix metalloproteinase-7 (MMP-7) were examined by immunohistochemistry in samples from 53 LSCC patients. Compared with normal laryngeal squamous epithelium (NLSE), there were upregulated expression of TC1, downregulated expression of Chibby, and aberrant cytoplasmic expression of β-catenin in the LSCC tissues (P < .001). The high expression of TC1 was correlated with the tumor site, advanced TNM and T stage, lymphovascular invasion, and poor differentiation in LSCC tissues (P < .050). There were correlations between the aberrant expression of β-catenin and the tumor site, advanced TNM and T stage, lymphovascular invasion, perineurial invasion, and poor differentiation in LSCC tissues (P < .050). Upregulated TC1 and downregulated Chibby were correlated with aberrant expression of β-catenin (P < .001), but no correlation between them (P = .076). The percent of abnormal expression of β-catenin in LSCC was 96.00% in TC1+/Chibby−, 73.68% in TC1+/Chibby+, 0.00% in TC1-/Chibby−, and 0.00% in TC1-/Chibby + group (P < .001). High expression of c-Myc, Cyclin D1, and MMP-7 was observed in LSCC tissues (P < .001). There was statistically significant about the expression of Cyclin D1 and MMP-7 among the groups of TC1+/Chibby−, TC1+/Chibby+, TC1-/Chibby−, and TC1-/Chibby + (P < .001), but was not significance about the expression of c-Myc among them (P = .339). No association was found between overall survival and the expression of TC1, Chibby, and β-catenin (P > .05). The upregulated expression of TC1 and downregulated expression of Chibby were correlated with the aberrant expression of β-catenin and the high expression of Cyclin D1 and MMP-7 in LSCC tissues.
... Moreover, they also had marked increases in Wnt target genes, including Axin2, C-myc and Ccnd1 (refs. [43][44][45]. Notably, these results are consistent with previous findings, in which recombinant SFRP2 was found to induce melanin synthesis in human primary melanocytes, via Wnt signalling activation to promote darkening of skin explants 19 . ...
Article
Full-text available
Animal pigment patterns are excellent models to elucidate mechanisms of biological organization. Although theoretical simulations, such as Turing reaction–diffusion systems, recapitulate many animal patterns, they are insufficient to account for those showing a high degree of spatial organization and reproducibility. Here, we study the coat of the African striped mouse (Rhabdomys pumilio) to uncover how periodic stripes form. Combining transcriptomics, mathematical modelling and mouse transgenics, we show that the Wnt modulator Sfrp2 regulates the distribution of hair follicles and establishes an embryonic prepattern that foreshadows pigment stripes. Moreover, by developing in vivo gene editing in striped mice, we find that Sfrp2 knockout is sufficient to alter the stripe pattern. Strikingly, mutants exhibited changes in pigmentation, revealing that Sfrp2 also regulates hair colour. Lastly, through evolutionary analyses, we find that striped mice have evolved lineage-specific changes in regulatory elements surrounding Sfrp2, many of which may be implicated in modulating the expression of this gene. Altogether, our results show that a single factor controls coat pattern formation by acting both as an orienting signalling mechanism and a modulator of pigmentation. More broadly, our work provides insights into how spatial patterns are established in developing embryos and the mechanisms by which phenotypic novelty originates.
... Morphologically, WNT/β-catenin-mutated HCC tumors usually exhibit well-differentiated, micro-beam skeleton, pseudo-glandular architecture pattern, tumor cholestasis, and lack of inflammatory infiltration phenotypes (Calderaro et al., 2019). As a part of the underlying mechanism, several oncoproteins in HCC have been characterized as targets of β-catenin, such as c-Myc (He et al., 1998), Cyclin D1 (Shtutman et al., 1999), TBX3 (Liang et al., 2021), and KIF2C (Wei et al., 2021), which are implicated in HCC progression, metastasis, metabolism, and drug resistance (Khalaf et al., 2018). It is worthy noticing that β-catenin mutation also functions in HCC metabolism, as Glutamine synthetase (GS, or Glutamate-ammonia ligase, GLUL) in glutaminolysis is the target of β-catenin (Cadoret et al., 2002), and its expression is correlated with β-catenin GOF mutation in HCC . ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is a subtype of highly malignant carcinoma that occurs in the liver, improved understanding of the mechanisms behind HCC tumorigenesis and better clinical treatment options are urgently needed. Several pieces of evidence have implied that the tumorigenesis and progression of HCC are driven by various genomic mutations and alterations. In this review, we have provided an overview of driver mutations in different signaling pathways that dominate HCC tumorigenesis, as well as vital molecular events in HCC initiation. Meanwhile, we have also summarized different agents or tools that may be utilized for HCC treatment in patients with corresponding mutation events. These findings may expand our understanding of the inherent characteristics of HCC and provide new perspectives for the future clinical treatment of HCC.
... HFDPCs were seeded in 96-well plates at a density of 1 × 10 4 ...
Article
Full-text available
Background When Wnt binds to the N‐terminal of Frizzled, a conformational change occurs in the C‐terminal of Frizzled, which binds to Dishevelled1 (Dvl1), a Wnt signaling component protein. When Dvl1 binds to the C‐terminal of Frizzled, the concentration of β‐catenin increases and it enters the nucleus to transmit cell proliferation signals. CXXC‐type zinc finger protein 5 (CXXC5) binds to the Frizzled binding site of Dvl1 and interferes with Dvl1–Frizzled binding. Therefore, blocking CXXC5–Dvl1 binding may induce Wnt signal transduction. Materials and methods We used WD‐aptamer, a DNA aptamer that specifically binds to Dvl1 and interferes with CXXC5–Dvl1 interaction. We confirmed the penetration of WD‐aptamer into human hair follicle dermal papilla cells (HFDPCs) and measured β‐catenin expression following treatment with WD‐aptamer in HFDPCs, wherein Wnt signaling was activated by Wnt3a. In addition, MTT assay was performed to investigate the effect of WD‐aptamer on cell proliferation. Results WD‐aptamer penetrated the cell, affected Wnt signaling, and increased β‐catenin expression, which plays an important role in signaling. Additionally, WD‐aptamer induced HFDPC proliferation. Conclusion CXXC5‐associated negative feedback of Wnt/β‐catenin signaling can be regulated by interfering with CXXC5–Dvl1 interaction.
... The binding of Wnt to its receptors suppresses the β-catenin destruction complex, composed of adenomatous polyposis coli (APC), Axin, casein kinase 1α (CK1α), GSK3β, and free β-catenin. The nuclear translation of β-catenin promotes the transcription of proliferationrelated genes [155][156][157]. Cytoplasmic β-catenin might form a complex with adherent junctions, promoting cell adhesion. ...
Article
Full-text available
Illnesses following the degeneration of the nervous system can occur due to aging or genetic mutations and represent a clinical concern. In neurodegenerative diseases, loss of neuronal structure and functions mainly causes cognitive impairment, representing an increasing social burden. In neurodegenerative diseases, the progressive loss of vulnerable populations of neurons in specific regions of the central nervous system was traced to different pathological events, such as misfolded proteins’ accumulation, abnormalities in proteasomes or phagosomes, as well as anomalies in lysosomes or mitochondria. Many research efforts identified important events involved in neurodegeneration, but the complex pathogenesis of neurodegenerative diseases is far from being fully elucidated. More recently, insights into the signal transduction pathways acting in the nervous system contributed to unveiling some molecular mechanisms triggering neurodegeneration. Abnormalities in the intra- or inter-cellular signaling were described to be involved in the pathogenesis of neurodegenerative disease. Understanding the signal transduction pathways that impact the nervous system homeostasis can offer a wide panel of potential targets for modulating therapeutic approaches. The present review will discuss the main signal transduction pathways involved in neurodegenerative disorders.
Article
Full-text available
Optimal lung repair and regeneration is essential for recovery from viral infections including influenza A virus (IAV). We have previously demonstrated that acute inflammation and mortality induced by IAV is under circadian control. However, it is not known if the influence of the circadian clock persists beyond the acute outcomes. Here, we utilize the UK Biobank to demonstrate an association between poor circadian rhythms and morbidity from lower respiratory tract infections including the need for hospitalization and post-discharge mortality; this persists even after adjusting for common confounding factors. Further, we use a combination of lung organoid assays, single cell RNA sequencing (Sc-seq) and IAV infection in different models of clock disruption to investigate the role of the circadian clock in lung repair and regeneration. We show for the first time that lung organoids have a functional circadian clock, and the disruption of this clock impairs regenerative capacity. Finally, we find that the circadian clock acts through distinct pathways in mediating lung regeneration- in tracheal cells via the Wnt/β-catenin pathway and through IL1β in alveolar epithelial cells. We speculate, that adding a circadian dimension to the critical process of lung repair and regeneration will lead to novel therapies and improve outcomes.
Chapter
Full-text available
Colorectal cancer is the cancer of the colon, located at the lower part of the digestive system. Although the role of STAT3 in cancer is known, its role particularly in colon cancer is largely unknown. STAT3 is a cytoplasmic transcription factor that involves extracellular signaling to the nucleus regulating fundamental functions, like cell proliferation, apoptosis, differentiation, and angiogenesis. STAT3 is a key regulator; abrogated activation leads to several diseases, including cancer. Aberrant interleukin (IL)-6-mediated JAK/STAT3 signaling pathway is closely related to the advancement of several human solid tumors including colorectal cancer. With other upstream regulators, IL-6/JAK signaling can activate STAT3, and its role appears to be critical in various types of cancer. STAT3 has been traditionally recognized as an oncogene; more recently the dual role of STAT3 in cancer, either tumor inductive or suppressive, has been appreciated. This chapter describes the potential role of STAT3 in colon cancer based on in vitro, in vivo, and patient studies. Furthermore, we will discuss the mechanism of action and roles of the IL-6/JAK/STAT3 pathway in colorectal cancer and exploit current therapeutic strategies, to treat colorectal cancer. Understanding the complexity of STAT3 function in colorectal cancer has the potential to elucidate important molecular aspects of colorectal cancer with significant therapeutic implications.
Article
Full-text available
β-Catenin and plakoglobin (γ-catenin) are closely related molecules of the armadillo family of proteins. They are localized at the submembrane plaques of cell–cell adherens junctions where they form independent complexes with classical cadherins and α-catenin to establish the link with the actin cytoskeleton. Plakoglobin is also found in a complex with desmosomal cadherins and is involved in anchoring intermediate filaments to desmosomal plaques. In addition to their role in junctional assembly, β-catenin has been shown to play an essential role in signal transduction by the Wnt pathway that results in its translocation into the nucleus. To study the relationship between plakoglobin expression and the level of β-catenin, and the localization of these proteins in the same cell, we employed two different tumor cell lines that express N-cadherin, and α- and β-catenin, but no plakoglobin or desmosomal components. Individual clones expressing various levels of plakoglobin were established by stable transfection. Plakoglobin overexpression resulted in a dose-dependent decrease in the level of β-catenin in each clone. Induction of plakoglobin expression increased the turnover of β-catenin without affecting RNA levels, suggesting posttranslational regulation of β-catenin. In plakoglobin overexpressing cells, both β-catenin and plakoglobin were localized at cell– cell junctions. Stable transfection of mutant plakoglobin molecules showed that deletion of the N-cadherin binding domain, but not the α-catenin binding domain, abolished β-catenin downregulation. Inhibition of the ubiquitin-proteasome pathway in plakoglobin overexpressing cells blocked the decrease in β-catenin levels and resulted in accumulation of both β-catenin and plakoglobin in the nucleus. These results suggest that (a) plakoglobin substitutes effectively with β-catenin for association with N-cadherin in adherens junctions, (b) extrajunctional β-catenin is rapidly degraded by the proteasome-ubiquitin system but, (c) excess β-catenin and plakoglobin translocate into the nucleus.
Article
Full-text available
Several different oncogenes and growth factors promote G1 phase progression. Cyclin D1, the regulatory subunit of several cyclin-dependent kinases, is required for, and capable of shortening, the G1 phase of the cell cycle. The present study demonstrates that transforming mutants of p21 (Ras Val-12, Ras Leu-61) induce the cyclin D1 promoter in human trophoblasts (JEG-3), mink lung epithelial (Mv1.Lu), and in Chinese hamster ovary fibroblast cell lines. Site-directed mutagenesis of AP-1-like sequences at −954 abolished p21-dependent activation of cyclin D1 expression. The AP-1-like sequences were also required for activation of the cyclin D1 promoter by c-Jun. In electrophoretic mobility shift assays using nuclear extracts from cultured cells and primary tissues, several AP-1 proteins (c-Jun, JunB, JunD, and c-Fos) bound the cyclin D1 −954 region. Cyclin D1 promoter activity was stimulated by overexpression of mitogen-activated protein kinase (p41) or c-Ets-2 through the proximal 22 base pairs. Expression of plasmids encoding either dominant negative MAPK (p41) or dominant negatives of ETS activation (Ets-LacZ), antagonized MAPK-dependent induction of cyclin D1 promoter activity. Epidermal growth factor induction of cyclin D1 transcription, through the proximal promoter region, was antagonized by either p41 or Ets-LacZ, suggesting that ETS functions downstream of epidermal growth factor and MAPK in the context of the cyclin D1 promoter. The activation of cyclin D1 transcription by p21provides evidence for cross-talk between the p21 and cell cycle regulatory pathways.
Article
Full-text available
The integrin-linked kinase (ILK) is an ankyrin repeat containing serine-threonine protein kinase that can interact directly with the cytoplasmic domains of the β1 and β3 integrin subunits and whose kinase activity is modulated by cell–extracellular matrix interactions. Overexpression of constitutively active ILK results in loss of cell–cell adhesion, anchorage-independent growth, and tumorigenicity in nude mice. We now show that modest overexpression of ILK in intestinal epithelial cells as well as in mammary epithelial cells results in an invasive phenotype concomitant with a down-regulation of E-cadherin expression, translocation of β-catenin to the nucleus, formation of a complex between β-catenin and the high mobility group transcription factor, LEF-1, and transcriptional activation by this LEF-1/β-catenin complex. We also find that LEF-1 protein expression is rapidly modulated by cell detachment from the extracellular matrix, and that LEF-1 protein levels are constitutively up-regulated at ILK overexpression. These effects are specific for ILK, because transformation by activated H-ras or v-src oncogenes do not result in the activation of LEF-1/β-catenin. The results demonstrate that the oncogenic properties of ILK involve activation of the LEF-1/β-catenin signaling pathway, and also suggest ILK-mediated cross-talk between cell–matrix interactions and cell–cell adhesion as well as components of the Wnt signaling pathway.
Article
Full-text available
The APC tumor-suppressor protein associates with beta-catenin, a cell adhesion protein that is upregulated by the WNT1 oncogene. We examined the effects of exogenous APC expression on the distribution and amount of beta-catenin in a colorectal cancer cell containing only mutant APC. Expression of wild-type APC caused a pronounced reduction in total beta-catenin levels by eliminating an excessive supply of cytoplasmic beta-catenin indigenous to the SW480 colorectal cancer cell line. This reduction was due to an enhanced rate of beta-catenin protein degradation. Truncated mutant APC proteins, characteristic of those associated with cancer, lacked this activity. Mutational analysis revealed that the central region of the APC protein, which is typically deleted or severely truncated in tumors, was responsible for the down-regulation of beta-catenin. These results suggest that the tumor-suppressor activity of mutant APC may be compromised due to a defect in its ability to regulate beta-catenin.
Article
Inactivation of the adenomatous polyposis coli (APC) tumor suppressor protein is responsible for both inherited and sporadic forms of colon cancer. Growth control by APC may relate to its ability to downregulate beta-catenin post-translationally. In cancer, mutations in APC ablate its ability to regulate beta-catenin, and mutations in beta-catenin prevent its downregulation by wild-type APC. Moreover, signaling by the protein product of the wnt-1 proto-oncogene upregulates beta-catenin and promotes tumorigenesis in mice. In a Xenopus developmental system, Wnt-1 signaling was inhibited by Axin, the product of the murine fused gene. This suggests a possible link between Axin, the Wnt-1 signaling components beta-catenin and glycogen synthase kinase 3 beta (GSK3 beta), and APC. Human Axin (hAxin) binds directly to beta-catenin, GSK3 beta, and APC in vitro, and the endogenous proteins are found in a complex in cells. Binding sites for Axin were mapped to a region of APC that is typically deleted due to cancer-associated mutations in the APC gene. Overexpression of hAxin strongly promoted the downregulation of wild-type beta-catenin in colon cancer cells, whereas mutant oncogenic beta-catenin was unaffected. The downregulation was increased by deletion of the APC-binding domain from Axin, suggesting that APC may function to derepress Axin activity. In addition, hAxin dramatically facilitated the phosphorylation of APC and beta-catenin by GSK3 beta in vitro. Axin acts as a scaffold upon which APC, beta-catenin and GSK3 beta assemble to coordinate the regulation of beta-catenin signaling.
Article
A protein first identified by its association with cadherin cell adhesion molecules, β-catenin, has been implicated in carcinogenesis. In a number of different types of cancer, signalling through β-catenin is upregulated either by direct mutation of β-catenin or loss of negative regulation by the APC tumor suppressor protein.
Article
Beta-catenin is a pivotal player in the signaling pathway initiated by Wnt proteins, mediators of several developmental processes. beta-catenin activity is controlled by a large number of binding partners that affect the stability and the localization of beta-catenin and is thereby able to participate in such varying processes as gene expression and cell adhesion. Activating mutations in beta-catenin and in components regulating its stability can contribute to the formation of certain tumors.
Article
Coordinated interactions between cyclin-dependent kinases (Cdks), their target “pocket proteins” (the retinoblastoma protein [pRB], p107, and p130), the pocket protein binding E2F-DP complexes, and the Cdk inhibitors regulate orderly cell cycle progression. The cyclin D1 gene encodes a regulatory subunit of the Cdk holoenzymes, which phosphorylate the tumor suppressor pRB, leading to the release of free E2F-1. Overexpression of E2F-1 can induce apoptosis and may either promote or inhibit cellular proliferation, depending upon the cell type. In these studies overexpression of E2F-1 inhibited cyclin D1-dependent kinase activity, cyclin D1 protein levels, and promoter activity. The DNA binding domain, the pRB pocket binding region, and the amino-terminal Sp1 binding domain of E2F-1 were required for full repression of cyclin D1. Overexpression of pRB activated the cyclin D1 promoter, and a dominant interfering pRB mutant was defective in cyclin D1 promoter activation. Two regions of the cyclin D1 promoter were required for full E2F-1-dependent repression. The region proximal to the transcription initiation site at −127 bound Sp1, Sp3, and Sp4, and the distal region at −143 bound E2F-4–DP-1–p107. In contrast with E2F-1, E2F-4 induced cyclin D1 promoter activity. Differential regulation of the cyclin D1 promoter by E2F-1 and E2F-4 suggests that E2Fs may serve distinguishable functions during cell cycle progression. Inhibition of cyclin D1 abundance by E2F-1 may contribute to an autoregulatory feedback loop to reduce pRB phosphorylation and E2F-1 levels in the cell.