Download Cell regulation by the Apc protein Apc as master regulator of epithelia

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Hedgehog signaling pathway wikipedia , lookup

Cell nucleus wikipedia , lookup

Cell encapsulation wikipedia , lookup

Apoptosis wikipedia , lookup

Endomembrane system wikipedia , lookup

Cell culture wikipedia , lookup

Cell growth wikipedia , lookup

Phosphorylation wikipedia , lookup

Tissue engineering wikipedia , lookup

Cell cycle wikipedia , lookup

Cellular differentiation wikipedia , lookup

Extracellular matrix wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Mitosis wikipedia , lookup

Protein phosphorylation wikipedia , lookup

Signal transduction wikipedia , lookup

Apoptosome wikipedia , lookup

Cytokinesis wikipedia , lookup

Spindle checkpoint wikipedia , lookup

Amitosis wikipedia , lookup

Biochemical switches in the cell cycle wikipedia , lookup

Secreted frizzled-related protein 1 wikipedia , lookup

List of types of proteins wikipedia , lookup

Transcript
Available online at www.sciencedirect.com
Cell regulation by the Apc protein
Apc as master regulator of epithelia
Brooke M McCartney1 and Inke S Näthke2
The adenomatous polyposis coli (Apc) protein participates in
many of the fundamental cellular processes that govern
epithelial tissues: Apc is directly involved in regulating the
availability of b-catenin for transcriptional de-repression of Tcf/
LEF transcription factors, it contributes to the stability of
microtubules in interphase and mitosis, and has an impact on
the dynamics of F-actin. Thus Apc contributes directly and/or
indirectly to proliferation, differentiation, migration, and
apoptosis. This particular multifunctionality can explain why
disruption of Apc is especially detrimental for the epithelium of
the gut, where Apc mutations are common in most cancers. We
summarise recent data that shed light on the molecular
mechanisms involved in the different functions of Apc.
Addresses
1
Department of Biological Sciences, Carnegie Mellon University, 4400
5th Avenue, Pittsburgh, PA, USA
2
College of Life Sciences, Cell & Dev. Biology, University of Dundee,
Dow Street, Dundee, Scotland, UK
Corresponding author: Näthke, Inke S ([email protected])
Current Opinion in Cell Biology 2008, 20:186–193
This review comes from a themed issue on
Cell regulation
Edited by Alan Hall and Joan Massagué
Available online 24th March 2008
0955-0674/$ – see front matter
Published by Elsevier Ltd.
DOI 10.1016/j.ceb.2008.02.001
The initial identification of the adenomatous polyposis coli
gene as the site of mutations in the familial form of colon
cancer, familial adenomatous polyposis (FAP), was
described in 1992 [1,2]. A causal relationship between
Apc mutations and intestinal tract tumours was confirmed
three years later with the establishment of the Min mouse
model [3]. These mice are heterozygous for Apc and
develop numerous intestinal tumours that mimic FAP.
Subsequently, Apc has emerged as the most commonly
mutated gene in colorectal cancer with reports of between
50 and >80% of sporadic tumours carrying such
mutations. The search for how mutations in Apc initiate
and/or support progression of tumours in the intestinal
tract revealed that Apc is a multifunctional participant in a
diverse array of cellular functions. In this review, we
describe these diverse functions and their relationship
to epithelial biology, and discuss recent significant
Current Opinion in Cell Biology 2008, 20:186–193
advances in our understanding of their underlying molecular mechanisms.
Wnt pathway regulation
The first recognised function of Apc was its role in Wnt
signalling [4,5]. This function is one of the driving
forces for how mutations in Apc ensure that cells remain
proliferative. Many of the molecular details of this pathway have been described extensively in many reviews [6].
Apc negatively regulates Wnt signalling by participating
in the destruction complex, a complex that targets the key
effector b-catenin for degradation (reviewed in [7]).
However, the precise role of Apc in the destruction
complex has not been clear. A number of recent papers
employing largely biochemical and structural approaches
have shed significant light on this question. The central
region of the Apc protein contains three distinct types of
repeats that are essential to its role in the negative
regulation of Wnt signalling (Figure 1). Seven 20 amino
acid repeats (20R) and three 15 amino acid repeats (15R)
function in the binding and degradation of b-catenin,
while three SAMP repeats bind to the RGS domain of
Axin, a partner in the destruction complex (reviewed [7]).
It has long been appreciated that phosphorylation of this
central repeat region of APC significantly enhances its
affinity for b-catenin [8]. The degree of conservation
between repeats of the same type suggested that they
might be functionally equivalent. However, recent work
has clearly demonstrated that individual 15R and 20R
differ significantly in their binding affinities for b-catenin
[9]. This difference seems to be achieved in part by the
N-terminal flanking regions of the repeats [9]. The 20R
with the highest binding affinity to b-catenin (R3, 4 and 5)
also contain a typical Tcf-like nine amino acid b-cateninbinding motif in their N-terminal flanking regions. The
Tcf/LEF family of transcription factors partner with bcatenin in the nucleus to activate Wnt target genes.
Changing a key aspartate residue in the flanking region
to alanine resulted in a 30-fold reduction in the binding
affinity for b-catenin. When the repeats are phosphorylated by the cooperative action of CK1 and GSK3b, the
binding interaction is significantly altered and enhanced.
Phosphorylation of any of the 20R, even those with no
detectable binding to b-catenin in a non-phosphorylated
state, increased their affinity for b-catenin dramatically.
When phosphorylated, binding of the 20R to b-catenin
was no longer modulated by the key aspartate in the Nterminal flanking region. Although the phosphorylation
sites of Apc have not been identified in vivo, the fact that
CK1 and GSK3b are endogenous components of the
www.sciencedirect.com
Regulation of cellular functions by the Apc protein McCartney and Näthke 187
Figure 1
Linear sequence schematic of Apc.The human Apc protein with 2843 amino acids is shown with the location of different functional sites.
destruction complex suggests that the CK1 and GSK3b
phosphorylation sites identified in vitro may be physiologically relevant [9].
These data significantly extend recently reported structural studies of the interaction between b-catenin and the
20R of Apc [10,11]. These studies demonstrated that
when phosphorylated, 20R3 and its N-terminal and Cterminal flanking sequences make contact with the entire
structural-binding groove of b-catenin. This contrasted
significantly with binding of unphosphorylated 20R3
[10]. Phosphorylated Apc can compete with Axin for
binding to b-catenin [10,12], but these proteins can
bind simultaneously when Apc is not phosphoryated
[10]. These observations suggest a model where phosphorylation of Apc is important for the recruitment and
turnover of b-catenin in the destruction complex. Phosphorylation of Apc binds Axin and b-catenin simultaneously, helping to bring these components together
(Figure 2). Once complexed, dephosphorylation of Apc
reduces its affinity for b-catenin, and b-catenin is transferred to Axin, but retains a reduced interaction with Apc.
Figure 2
Cycling of Apc phosphorylation may drive destruction complex function.(1) Apc with phosphorylated 20Rs has high affinity for b-catenin and may
facilitate the entry of b-catenin into the complex (2). (3) Dephosphorylation of Apc, perhaps by PP2A, drives the hand-off of b-catenin to Axin where it
can be phosphorylated by GSK3b. (4) P-b-catenin is identified by b-TrCP, the F-box subunit of the E3 ubiquitin ligase, ubiquitinated and degraded by
the proteosome. Modified from [7].
www.sciencedirect.com
Current Opinion in Cell Biology 2008, 20:186–193
188 Cell regulation
b-catenin phosphorylated by Axin-associated GSK3b is
targeted for ubiquitination and degradation by the proteosome. It is tempting to speculate that the phosphatase
responsible for dephosphorylating Apc is PP2A. This
phosphatase associates with Axin [13–15] and its regulatory subunit B56 interacts with both Axin and Apc through
its Arm repeats [16,17]. Consistent with this model,
mutations that disrupt the Arm repeats of Drosophila
APC2 also disrupt the function of the destruction complex [18]. Ha et al. tested this model by exposing the
phosphorylated-Apc/b-catenin complex to PP1, a biochemically tractable phosphatase similar to PP2A.
Although PP1 could effectively dephosphorylate Apc
alone, Apc complexed with b-catenin was resistant to
its activity [10]. Although PP2A within the destruction
complex may function differently, the mechanisms
for dephosphorylating Apc within the complex clearly
require more careful examination. Cycling of Apc
phosphorylation has also been suggested to affect the
release of b-catenin from the complex [12]. However,
the presence of Apc in a complex with b-catenin and the
ubiquitin ligase subunit b-TrCP [19] suggests that complete release from the destruction complex may not be
necessary to hand over phosphorylated b-catenin to the
proteosome [10] (Figure 2).
Although many details of this pathway remain unknown,
it is clear that phosphorylation of the 20R is a key step in
regulating the binding of Apc to b-catenin. Furthermore,
the different binding properties of each repeat suggest
that they may play different roles in distinct cellular
contexts [9]. In the unphosphorylated state, the binding affinities of any of the 20R except 20R3 are likely too
low to be relevant. The different binding affinities in the
phosphorylated state may be important to fine tune
the Apc/b-catenin interactions that occur not only in
the destruction complex, but also at other sites in the
cell including the nucleus (see below), the cortex [20,21],
and at the centrosome [22]. Further dissection of the
repeats using a wide range of functional assays will be an
essential step to understand this interaction. In the Wnt
signalling context, identification of 20R3 as the highest
affinity b-catenin binding repeat provides a more
detailed explanation for the observation that the
mutation cluster region (between amino acids 1250
and 1450 (Figure 1)) is associated with human colon
cancer. Truncated proteins resulting from these
mutations lack all of the Axin-binding SAMP repeats
as well as 20R3–7, retaining only two of the weakest
binding repeats 20R1-2.
Nuclear Apc
Phosphorylation of Apc is emerging as a key mechanism
for regulating Apc function in the canonical destruction
complex as well as in the regulation of Apc’s subcellular
distribution. As part of its role as a negative regulator of
Wnt signalling, Apc proteins shuttle in and out of the
Current Opinion in Cell Biology 2008, 20:186–193
nucleus where they interact with b-catenin, CtBP, and
other proteins involved in transcription (reviewed in
[23]). Consistent with these observations, Apc contains
two classic monopartite basic nuclear localisation signals
in the C-terminal half of the protein and an additional
domain within the Arm repeats that facilitates nuclear
import (Figure 1). The regulation of Apc’s nuclear import
is complex and involves a number of factors including the
activity of several NESs (Figure 1), sequestration of Apc
at non-nuclear sites by interactions with other protein
partners, and regulation of NLS activity (reviewed in
[23]). Regulation of NLS activity involves, at least in
part, the phosphorylation of NLS flanking sites where
phosphorylation by casein kinase 2 (CK2) may enhance
NLS activity, while phosphorylation by protein kinase A
(PKA) inhibits NLS activity [24]. Recent evidence
suggests that p38 MAPK and its regulator Gadd45a act
to enhance the nuclear import of Apc by enhancing the
activity of CK2 and suppressing the activity of PKA [25].
The relationship between Apc and CK2 is complicated
not only does CK2 appear to phosphorylate Apc but the
direct binding of Apc to CK2 inhibits CK2 activity [26],
suggesting that Apc imposes negative feedback on its own
nuclear localisation.
What is the function of nuclear Apc? Mounting evidence
favours the hypothesis that Apc sequesters nuclear bcatenin. This is supported by careful observations of
nucleo-cytoplasmic shuttling of b-catenin using live cell
microscopy and fluorescence recovery after photobleaching (FRAP) [27], which revealed that none of the factors
that bind b-catenin in the nucleus, including positive
effectors TCF4, BCL9/Pygopus and negative regulators
Apc and Axin influence the rate of b-catenin nuclear
import and export. Rather, the data suggest that these
proteins affect the compartmentalisation of b-catenin by
sequestering it at specific sites. Taken together, these
data support a model whereby p38 MAPK signalling
activates CK2 to phosphorylate Apc, promoting its
nuclear import where it acts to sequester b-catenin,
perhaps in cooperation with CtBP [28], and so downregulates b-catenin activity. An interesting twist to this
model was revealed by the finding that CK2 phosphorylates LEF1, and is required for the assembly and turnover of b-catenin/LEF1 nuclear complexes [29,30].
However, nuclear Apc may not be the only pool of Apc
that regulates nuclear b-catenin. A recent study of C.
elegans Apc, APR-1, suggests that cortical APR-1 functions
to regulate the Wnt pathway primarily by facilitating the
nuclear export of WRM1, a worm b-catenin [31]. Furthermore, although there has been significant progress in
understanding the mechanisms that regulate nuclear
Apc, its function in this compartment is not clear. There
are many ideas that have not been explored in detail,
particularly the possibilities that Apc itself may interact
with DNA [32], and components of the base excision
repair pathway [33].
www.sciencedirect.com
Regulation of cellular functions by the Apc protein McCartney and Näthke 189
Figure 3
Apc in intestinal tissue.(a) In wild-type intestinal tissue, stem cells near the base of the crypt (red), give rise to transit amplifying cells (yellow) that
differentiate into absorptive epithelial cells (green), neuroendocrine (brown), Paneth (blue), or Goblet (pink) cells. Differentiation is accompanied by
active migration along the crypt-villus axis. Near the top of villi in the small intestine (or in the collar of the crypt in the colon, where villi are not present),
cells are extruded and shed into the gut lumen while undergoing apoptosis (dark green). Apc contributes to this tissue organisation by supporting
differentiation, thus keeping proliferation restricted to the appropriate compartment, and migration to ensure continuous efflux of cells from the
proliferative compartment. It also contributes to apoptosis, and can enhance or inhibit cell death depending on the cellular context (position, tissue
type etc.). (b) In the absence of Apc, or when only mutated Apc is present, cells do not differentiate normally; they remain proliferative and retain some
stem cell characteristics (red-yellow). They also fail to migrate normally. Thus cells accumulate and form pre-malignant polyps that can deteriorate into
adenoma and carcinoma. The top of villi retains normal enterocytes, because each villus is supplied with new cells by many crypts and usually only one
crypt becomes malignant.
Apc in apoptosis
Mutations in Apc may also promote changes in apoptosis,
a crucial component of epithelial biology in the intestinal
tract. When cells reach the luminal collar of the colonic
crypt or the tip of the intestinal villus they are extruded
from the tissue while undergoing apoptosis and are shed
into the lumen of the gut (Figure 3) [34]. Although the
molecular mechanism for a role for Apc in apoptosis has
not been identified, evidence for a role of Apc in the
normal execution of an apoptotic program is accumulating
(reviewed in [35]). Whether components of the cell death
pathway are direct targets of Wnt signalling has yet to be
elucidated, though screens for Wnt target genes have
detected changes in genes encoding proteins directly
involved in the execution of apoptosis [36,37]. However,
other changes induced by Apc mutations (see below) may
also contribute [38]. Importantly, the overall balance of
pro-apoptotic and anti-apoptotic changes induced by Apc
loss may be highly context- and tissue-specific and may
cause increased or decreased apoptosis, depending on
tissue type, tissue compartment, developmental stages,
etc.
www.sciencedirect.com
Apc as a cytoskeletal regulator important in
migration, orientation and division
The transcriptional changes induced by mutant Apc provide the background against which the other cellular
functions of Apc become particularly important in the
digestive tract epithelium. In addition to its function in
the b-catenin destruction complex, Apc can also directly
contribute to the regulation of cytoskeletal proteins: it
binds directly and indirectly to microtubules and also
interacts with a number of actin-regulatory proteins
(Figures 1 and 4). So it is not surprising that loss of Apc
leads to changes in cell migration, cell orientation, polarity
and division [38,39,40,41]. Transcriptional changes
induced by Apc mutations also contribute to these defects;
nonetheless, direct interactions of Apc with cytoskeletal
elements contribute significantly and directly.
The stabilising effect on microtubules by Apc has been
established biochemically [42,43] and in cells [39,44]
and may be affected by the interaction of Apc with the
microtubule plus tip-binding protein EB1 [45]. The ability of C-terminal fragments of Apc to nucleate the assembly
Current Opinion in Cell Biology 2008, 20:186–193
190 Cell regulation
Figure 4
Figure 5
Apc complexes in migrating cellular protrusions. A number of proteins
support the linkage between Apc and microtubules, but also F-actin in
migrating cellular protrusions near the plasma membrane. Whether all of
these interactions can occur at the same time, and how they affect each
other is not known.
Apc in mitotic spindles.Apc (red dot) localises to spindle microtubules,
concentrates at kinetochores, centrosomes and at sites of cortical
attachment. At these different sites Apc may co-operate with the
indicated proteins to support microtubule attachment and dynamics at
both kinetochores and cortical sites, but also at centrosomes.
of F-actin in vitro [46] together with the ability of Apc to
interact with a number of actin-regulatory proteins including ASEF, an exchange factor for CDC42 and Rac,
and IQGAP, suggests that Apc also contributes to the
regulation of the cellular F-actin network [47,48]. The
idea that Apc helps to co-ordinate the F-actin and microtubule cytoskeletons is further supported by the interaction of Apc with mDia, a protein that can affect both
actin and microtubules [49] (Figure 4). How these interactions are co-ordinated, whether they occur simultaneously or whether these proteins compete with each
other, and how they contribute individually or together to
the localised recruitment of Apc clusters in migrating
cellular protrusion is not at all clear and will require careful
biochemical and functional experiments (Figure 4).
function of the microtubule-based mitotic spindle
(Figure 5). Loss of Apc leads to defects in spindle
alignment [51], and the mitotic spindle checkpoint
[38], and to the development of anaphase bridges
[52]. An immediate consequence of these changes is
the accumulation of tetraploid cells, not only in culture,
but also in intestinal tissue depleted of Apc [38]. Similar
defects are observed in cells expressing N-terminal fragments of Apc [53] and intestinal tissue from Min mice
displays failures in cytokinesis [54]. The expression of Nterminal fragments of Apc, similar to those present in
human tumours, may disrupt normal Apc function by
forming complexes with wild-type Apc and interfering
with its normal interactions [55]. One possible interaction
that may be affected by such Apc heterodimeric complexes is the binding of Apc to EB1 [56].
Nonetheless, it has been clearly established that these
interactions contribute to a number of cellular processes
that are crucial for gut epithelium maintenance, most
prominently in cell migration. Differentiating epithelial
cells in the gut actively migrate upwards from the crypts
and loss of Apc leads to a decrease in this migration [50]
(Figure 3). Similarly, depleting Apc from cultured cells
leads to a decrease in the ability to migrate in a directed
fashion [39] that is accompanied by a decrease in overall
microtubule stability and loss of polarisation of acetylated
microtubules towards the leading edge [39,44]. Similar
to migrating epithelial cells, highly polarised migrating
astrocytes also accumulate Apc at their leading edge and
loss of Apc causes a loss of migration in these cells [40].
The cytoskeletal interactions of Apc not only affect cells
in interphase but also contribute significantly to the
Current Opinion in Cell Biology 2008, 20:186–193
The interaction between Apc and EB1 and thus dynein/
dynactin, which binds to EB1 directly, may also contribute to the ability of Apc to support anchoring of astral
microtubules at cortical sites (Figure 5) [54,57–59]. Similarly this interaction may contribute to the function(s) of
Apc at the centrosome [22]. Together these functions of
Apc in the mitotic spindle could explain the spindle
positions defects in Apc mutant cells [53].
Apc in whole tissue
Much of the evidence for the functions of Apc is based on
data obtained in cultured cells. However, important
insights into how these functions contribute to tissue
homeostasis and tumourigenesis have been gained using
animal models. Mice with constitutive or conditional
mutations in Apc confirmed its contribution to differenwww.sciencedirect.com
Regulation of cellular functions by the Apc protein McCartney and Näthke 191
tiation, migration, division and genetic stability: both Min
mice and PIRC rats are heterozygous for Apc mutations,
similar to FAP patients, and both develop tumours in the
digestive tract similar to those in human FAP patients
[60]. Conditional depletion of Apc specifically in the
intestine results in major changes in cell differentiation,
proliferation, ploidy and apoptosis consistent with the
findings in cell culture systems [50]. Importantly, the
defects induced by acute loss of Apc in gut tissue do not
occur when c-myc is also missing [61]. Cells lacking both
of these genes are selectively depleted from intestinal
tissue within a few weeks. It is possible that a downstream
target of c-myc that is upregulated when Apc is lost is
responsible for the migration defect observed in Apcdepleted cells. One difficulty in measuring a change in
migration in c-myc-depleted cells in this context is that
these cells do not survive and are out-competed rapidly
by wild-type cells in this tissue. This means that mixed
populations of cells, plus and minus c-myc, co-exist and it
is impossible to resolve how mixing between such cells
affects migration and general cell behaviour. However,
during the lifetime of these double-mutant cells in intestinal tissue, b-catenin is nuclear suggesting that it is
acting as a transcriptional activator [61]. These data
suggest that upregulation of c-myc is absolutely required
for Apc-deficient cells to survive.
Conclusion
Many of the defects that result from Apc-deficiency can
be overcome by overexpressing other components of
the relevant pathways: overexpression of Axin can rescue b-catenin regulation in Apc-deficient cells [62], cell
cycle checkpoint deficiency can be rescued by overexpressing Bub1 [38]. Furthermore, Apc is not absolutely required for any of the processes discussed above
and most of them continue to proceed, albeit less
efficiently in its absence: cells still move, but more
slowly and not as directed; cells divide, but make more
mistakes in this process; apoptotic pathways still function, but are not initiated appropriately in response to
certain checkpoints [38]. In support of the notion that
Apc is not essential for these processes, Drosophila
completely lacking Apc do not exhibit detectable
defects in mitosis, cytokinesis, cell polarity, or adhesion,
but lethality appears to result primarily from excessive
Wnt signalling [18]. A role for Apc in ensuring
accuracy and efficiency of many cellular processes
explains why Apc mutations are particularly detrimental
for tissues that rely on the fidelity of these processes.
The cumulative destabilisation that results from Apc
disruption makes Apc a perfect tumour suppressor,
particularly in gut epithelial tissues: loss of Apc leads
to a large increase in mistakes in the processes that
govern the overall homeostasis of this tissue and these
mistakes are not corrected, instead they are tolerated
allowing these oncogenic changes to propagate.
www.sciencedirect.com
References and recommended reading
Papers of particular interest, published within the annual period of
review, have been highlighted as:
of special interest
of outstanding interest
1.
Su LK, Kinzler KW, Vogelstein B, Preisinger AC, Moser A,
Luongo C, Gould KA, Dove WF: Multiple intestinal neoplasia
caused by mutations in the murine homolog of the APC gene.
Science 1992, 256:668-670.
This paper identifies the Apc locus as the site for mutations in intestinal
neplasia.
2.
Moser AR, Dove WF, Roth KA, Gordon JI: The Min (multiple
intestinal neoplasia) mutation: its effect on gut epithelial cell
differentiation and interaction with a modifier system. J Cell
Biol 1992, 116:1517-1526.
This paper identifies the Apc locus as the site for mutations in intesitnal
neplasia, but also demonstrates the effects of genetic background on
penetrance of phenotype
3.
Moser AR, Luongo C, Gould KA, McNeley MK, Shoemaker AR,
Dove WF: ApcMin: a mouse model for intestinal and mammary
tumorigenesis. Eur J Cancer 1995, 31A:1061-1064.
This paper established the causal relationship between Apc heterozygosity and intestinal neoplasia. The Min mouse described in this paper is
one of the most commonly used mouse strains in the world.
4.
Rubinfeld B, Souza B, Albert I, Müller O, Chamberlain SH,
Masiarz FR, Munemitsu S, Polakis P: Association of the APC
gene product with b-catenin. Science 1993, 262:1731-1734.
This paper shows the interaction of Apc protein with b-catenin, which set
the stage for the identifying the Wnt-regulated degradation of b-catenin
as a central pathway in colon tumour development.
5.
Su L-K, Vogelstein B, Kinzler KW: Association of the APC tumor
suppressor protein with catenins. Science 1993, 262:17341737.
Shows the interaction of Apc protein with b-catenin, which set the stage
for the identifying the Wnt-regulated degradation of b-catenin as a central
pathway in colon tumour development.
6.
Polakis P: The many ways of Wnt in cancer. Curr Opin Genet Dev
2007, 17:45-51.
7.
Kennell J, Cadigan K: APC and ß-catenin degradation. In APC
proteins. Edited by Näthke IS, McCartney BM: Landes Bioscience;
2008, in press. http://www.eurekah.com/chapter/3776.
8.
Rubinfeld B, Albert I, Porfiri E, Fiol C, Munemitsu S, Polakis P:
Binding of GSK3beta to the APC-beta-catenin complex and
regulation of complex assembly. Science 1996, 272:1023-1026.
This paper provides significant insights into the assembly and regulation
of the complex that governs the degradation of b-catenin. Of particular
importance to this discussion was the finding that the phosphorylation of
the central repeat region of Apc significantly enhances its affinity for bcatenin.
9.
Liu J, Xing Y, Hinds TR, Zheng J, Xu W: The third 20 amino acid
repeat is the tightest binding site of APC for beta-catenin. J
Mol Biol 2006, 360:133-144.
The authors present a detailed analysis of the binding affinities of
individual phosphorylated and unphosphorylated 15R and 20R for bcatenin. They showed that individual repeats have distinct binding affinities, with 20R3 being the highest, and the phosphorylation increases the
binding affinities for all of them. Furthermore, specific amino acids in the
20R N-terminal flanking sequences significantly enhance the binding
affinities of unphosphorylated 20R for b-catenin. Further study is required
to determine the functional significance of these distinctions.
10. Ha NC, Tonozuka T, Stamos JL, Choi HJ, Weis WI: Mechanism of
phosphorylation-dependent binding of APC to beta-catenin
and its role in beta-catenin degradation. Mol Cell 2004, 15:511521.
See annotation to [11].
11. Xing Y, Clements WK, Le Trong I, Hinds TR, Stenkamp R,
Kimelman D, Xu W: Crystal structure of a beta-catenin/APC
complex reveals a critical role for APC phosphorylation in APC
function. Mol Cell 2004, 15:523-533.
This paper along with [10] reported the crystal structure of Apc 20R
complexed with b-catenin. These studies made the surprising observaCurrent Opinion in Cell Biology 2008, 20:186–193
192 Cell regulation
tion that when phosphorylated not only does the 20R itself interact with bcatenin, but N-terminal and C-terminal flanking regions make contact as
well. Ha et al. [10] demonstrated that phosphorylated Apc (P-Apc)
competes with Axin for binding to b-catenin, while non-P-Apc and Axin
can bind b-catenin simultaneously. These findings suggest a novel model
for interactions within the destruction complex. Phosphorylation may be a
‘critical switch’ for Apc outside of the destruction complex as well. Xing
et al. 2004 showed that P-Apc competes with Tcf binding to b-catenin,
and thus may provide an additional mechanism for the downregulation of
b-catenin transcription by Apc.
12. Xing Y, Clements WK, Kimelman D, Xu W: Crystal structure of a
beta-catenin/axin complex suggests a mechanism for the betacatenin destruction complex. Genes Dev 2003, 17:2753-2764.
This paper provided important structural insights into the nature of the bcatenin-Axin complex that set the stage for the later structural work.
13. Gao ZH, Seeling JM, Hill V, Yochum A, Virshup DM: Casein kinase I
phosphorylates and destabilizes the beta-catenin degradation
complex. Proc Natl Acad Sci U S A 2002, 99:1182-1187.
14. Ikeda S, Kishida M, Matsuura Y, Usui H, Kikuchi A: GSK-3betadependent phosphorylation of adenomatous polyposis coli
gene product can be modulated by beta-catenin and protein
phosphatase 2A complexed with Axin. Oncogene 2000, 19:537545.
15. Hsu W, Zeng L, Constantini F: Identification of a domain of Axin
that binds to the serin/threonine protein phosphatase 2A and a
self-binding domain. J Biol Chem 1999, 274:3439-3445.
16. Seeling J, Miller J, Gil R, Moon R, White R, Virshup D: Regulation
of beta-catenin signaling by the B56 subunit of protein
phosphatase 2A. Science 1999, 283:2089-2091.
This paper demonstrates the involvement of PP2A as an important
regulator of b-catenin degradation.
17. Yamamoto H, Hinoi T, Michiue T, Fukui A, Usui H, Janssens V, Van
Hoof C, Goris J, Asashima M, Kikuchi A: Inhibition of the Wnt
signaling pathway by the PR61 subunit of protein phosphatase
2A. J Biol Chem 2001, 276:26875-26882.
18. McCartney BM, Price MH, Webb RL, Hayden MA, Holot LM,
Zhou M, Bejsovec A, Peifer M: Testing hypotheses for the
functions of APC family proteins using null and truncation
alleles in Drosophila. Development 2006, 133:2407-2418.
The authors showed that Drosophila null for both Apc genes (Apc1 and
APC2) do not exhibit significant defects in a variety of processes in which
Apc has been implicated, including cell viability, cadherin-based adhesion, spindle morphology and orientation, or cytokinesis. Complete loss
of Drosophila Apc proteins does result in significant activation of Wnt
signalling throughout development. These findings are consistent with the
hypothesis that although Apc proteins may function in many cellular
processes, they are not strictly required for most of these processes,
but rather function in their fidelity and/or efficiency.
19. Hart M, Concordet JP, Lassot I, Albert I, del los Santos R,
Durand H, Perret C, Rubinfeld B, Margottin F, Benarous R et al.:
The F-box protein beta-TrCP associates with phosphorylated
beta-catenin and regulates its activity in the cell. Curr Biol
1999, 9:207-210.
20. McCartney BM, Dierick HA, Kirkpatrick C, Moline MM, Baas A,
Peifer M, Bejsovec A: Drosophila APC2 is a cytoskeletallyassociated protein that regulates Wnt signaling in the
embryonic epidermis. J Cell Biol 1999, 146:1303-1318.
21. Yu X, Waltzer L, Bienz M: A new Drosophila APC homologue
associated with adhesive zones of epithelial cells. Nat Cell Biol
1999, 1:144-151.
22. Louie RK, Bahmanyar S, Siemers KA, Votin V, Chang P, Stearns T,
Nelson WJ, Barth AI: Adenomatous polyposis coli and EB1
localize in close proximity of the mother centriole and EB1 is a
functional component of centrosomes. J Cell Sci 2004,
117:1117-1128.
23. Neufeld K: Nuclear APC. In APC proteins. Edited by Näthke IS,
McCartney BM: Landes Bioscience; 2008, in press. http://
www.eurekah.com/chapter/3835.
24. Zhang F, White RL, Neufeld KL: Phosphorylation near nuclear
localization signal regulates nuclear import of adenomatous
polyposis coli protein. Proc Natl Acad Sci U S A 2000, 97:1257712582.
Current Opinion in Cell Biology 2008, 20:186–193
This paper provided key evidence that the NLS activity of Apc is regulated
by the phosphorylation of NLS flanking sites in two different ways.
Phosphorylation by casein kinase 2 appears to enhance NLS activity,
while phosphorylation by protein kinase A, inhibits NLS activity.
25. Hildesheim J, Salvador JM, Hollander MC, Fornace AJ Jr: Casein
kinase 2- and protein kinase A-regulated adenomatous
polyposis coli and beta-catenin cellular localization is
dependent on p38 MAPK. J Biol Chem 2005, 280:17221-17226.
26. Homma MK, Li D, Krebs EG, Yuasa Y, Homma Y: Association and
regulation of casein kinase 2 activity by adenomatous
polyposis coli protein. Proc Natl Acad Sci U S A 2002, 99:59595964.
27. Krieghoff E, Behrens J, Mayr B: Nucleo-cytoplasmic distribution
of beta-catenin is regulated by retention. J Cell Sci 2006,
119:1453-1463.
Using live cell imaging and fluorescence recovery after photobleaching,
the authors demonstrated that none of the factors known to bind to bcatenin in the nucleus influence the rate of b-catenin nuclear import and
export. Instead, these binding partners may act to sequester b-catenin in
specific cellular compartments. This elegant study strongly supports the
model that one function of Apc in the nucleus is to sequester b-catenin
and thus downregulate its transcriptional activity.
28. Hamada F, Bienz M: The APC tumor suppressor binds to Cterminal binding protein to divert nuclear beta-catenin from
TCF. Dev Cell 2004, 7:677-685.
29. Wang S, Jones KA: CK2 controls the recruitment of Wnt
regulators to target genes in vivo. Curr Biol 2006, 16:2239-2244.
See annotation to [30].
30. Tapia JC, Torres VA, Rodriguez DA, Leyton L, Quest AF: Casein
kinase 2 (CK2) increases survivin expression via enhanced
beta-catenin-T cell factor/lymphoid enhancer binding factordependent transcription. Proc Natl Acad Sci U S A 2006,
103:15079-15084.
This paper along with [29] provide evidence for additional roles for casein
kinase 2 (CK2) in Wnt signalling. Not only does CK2 appear to regulate the
nuclear localisation of Apc (24), but it also plays a role in the assembly of
LEF1-b-catenin transcriptional complexes.
31. Mizumoto K, Sawa H: Cortical beta-catenin and APC regulate
asymmetric nuclear beta-catenin localization during
asymmetric cell division in C. elegans. Dev Cell 2007, 12:287299.
32. Deka J, Herter P, Sprenger-Haussels M, Koosch S, Franz D,
Muller KM, Kuhnen C, Hoffmann I, Muller O: The APC protein
binds to A/T rich DNA sequences. Oncogene 1999, 18:56545661.
33. Narayan S, Jaiswal AS, Balusu R: Tumor suppressor APC blocks
DNA polymerase beta-dependent strand displacement
synthesis during long patch but not short patch base excision
repair and increases sensitivity to methylmethane sulfonate. J
Biol Chem 2005, 280:6942-6949.
34. Bullen TF, Forrest S, Campbell F, Dodson AR, Hershman MJ,
Pritchard DM, Turner JR, Montrose MH, Watson AJ:
Characterization of epithelial cell shedding from human small
intestine. Lab Invest 2006, 86:1052-1063.
35. Benchabane H, Ahmed Y: The adenomatous polyposis coli
tumor suppressor and wnt signaling in the regulation of
apoptosis. In APC Proteins. Edited by Näthke IS, McCartney BM.
Landes Bioscience, vol 1; 2008, in press. http://
www.eurekah.com/chapter/3784.
36. Ioannidis V, Beermann F, Clevers H, Held W: The beta-catenin —
TCF-1 pathway ensures CD4(+)CD8(+) thymocyte survival. Nat
Immunol 2001, 2:691-697.
37. Huang M, Wang Y, Sun D, Zhu H, Yin Y, Zhang W, Yang S, Quan L,
Bai J, Wang S et al.: Identification of genes regulated by Wnt/
beta-catenin pathway and involved in apoptosis via
microarray analysis. BMC Cancer 2006, 6:221.
38. Dikovskaya D, Schiffmann D, Newton IP, Oakley A, Kroboth K,
Sansom O, Jamieson TJ, Meniel V, Clarke A, Näthke IS: Loss of
APC induces polyploidy as a result of a combination of defects
in mitosis and apoptosis. J Cell Biol 2007, 176:183-195.
This paper demonstrates immediate defects in cell cycle machinery,
including the spindle checkpoint, after the loss of Apc in cells and tissue.
www.sciencedirect.com
Regulation of cellular functions by the Apc protein McCartney and Näthke 193
39. Kroboth K, Newton IP, Kita K, Dikovskaya D, Zumbrunn J,
Waterman-Storer CM, Näthke IS: Lack of adenomatous
polyposis coli protein correlates with a decrease in cell
migration and overall changes in microtubule stability. Mol Biol
Cell 2007, 18:910-918.
This paper shows that Apc status correlates with over microtubule
stability in cells and affects cell migration.
40. Etienne-Manneville S, Hall A: Cdc42 regulates GSK-3beta and
adenomatous polyposis coli to control cell polarity. Nature
2003, 421:753-756.
This paper provides important link between small GTPases and Apc in
migrating protrusions.
41. Etienne-Manneville S, Manneville JB, Nicholls S, Ferenczi MA,
Hall A: Cdc42 and Par6-PKCzeta regulate the spatially
localized association of Dlg1 and APC to control cell
polarization. J Cell Biol 2005, 170:895-901.
42. Munemitsu S, Souza B, Müller O, Albert I, Rubinfeld B, Polakis P:
The APC gene product associates with microtubules in vivo
and promotes their assembly in vitro. Cancer Res 1994,
54:3676-3681.
This is the first demonstration that Apc can bind to microtubules.
43. Zumbrunn J, Inoshita K, Hyman AA, Näthke IS: Binding of the
adenomatous polyposis coli protein to microtubules
increases microtubule stability and is regulated by GSK3b
phosphorylation. Curr Biol 2001, 11:44-49.
This paper shows the direct effect of Apc on microtubule stability in vitro
and in cells and provides evidence that this function can be regulated by
the phosphorylation with GSK3b.
44. Kita K, Wittmann T, Näthke IS, Waterman-Storer CM: APC on
microtubule plus ends in cell extensions can promote
microtubule net growth with or without EB1. Mol Biol Cell 2006,
17:2331-2345.
This paper demonstrates direct link between endogenous Apc protein
and microtubule dynamics in cells.
45. Su L-K, Burrell M, Hill DE, Gyuris J, Brent R, Wiltshire R, Trent J,
Vogelstein B, Kinzler KW: APC binds to the novel protein EB1.
Cancer Res 1995, 55:2972-2977.
This paper identifies EB1 as an Apc binding partner and thus brings Apc
closer to the microtubule cytoskeleton.
46. Moseley JB, Bartolini F, Okada K, Wen Y, Gundersen GG,
Goode BL: Regulated binding of adenomatous polyposis coli
protein to actin. J Biol Chem 2007, 282:12661-12668.
47. Kawasaki Y, Senda T, Ishidate T, Koyama R, Morishita T,
Iwayama Y, Higuchi O, Akiyama T: Asef, a link between the
tumor suppressor APC and G-protein signaling. Science 2000,
289:1194-1197.
Identifies Apc as a regulator for the activity of an exchange factor for small
GTP-ases thus establishing a functional link between Apc and actin.
48. Watanabe T, Wang S, Noritake J, Sato K, Fukata M, Takefuji M,
Nakagawa M, Izumi N, Akiyama T, Kaibuchi K: Interaction with
IQGAP1 links APC to Rac1, Cdc42, and actin filaments during
cell polarization and migration. Dev Cell 2004, 7:871-883.
49. Wen Y, Eng CH, Schmoranzer J, Cabrera-Poch N, Morris EJ,
Chen M, Wallar BJ, Alberts AS, Gundersen GG: EB1 and APC bind
to mDia to stabilize microtubules downstream of Rho and
promote cell migration. Nat Cell Biol 2004, 6:820-830.
50. Sansom OJ, Reed KR, Hayes AJ, Ireland H, Brinkmann H,
Newton IP, Batlle E, Simon-Assmann P, Clevers H, Näthke IS
et al.: Loss of Apc in vivo immediately perturbs Wnt
www.sciencedirect.com
signaling, differentiation, and migration. Genes Dev 2004,
18:1385-1390.
Shows that conditionally inactivating Apc in the gut immediately leads to
changes that underlie tumourigenesis.
51. Draviam VM, Shapiro I, Aldridge B, Sorger PK: Misorientation and
reduced stretching of aligned sister kinetochores promote
chromosome missegregation in EB1- or APC-depleted cells.
EMBO J 2006, 25:2814-2827.
52. Aoki K, Aoki M, Sugai M, Harada N, Miyoshi H, Tsukamoto T,
Mizoshita T, Tatematsu M, Seno H, Chiba T et al.: Chromosomal
instability by beta-catenin/TCF transcription in APC or betacatenin mutant cells. Oncogene 2007, 26:3511-3520.
53. Green RA, Kaplan KB: Chromosome instability in colorectal
tumor cells is associated with defects in microtubule plus-end
attachments caused by a dominant mutation in APC. J Cell Biol
2003, 163:949-961.
This paper demonstrates a dominant effect of N-terminal Apc fragments
in mitosis.
54. Caldwell CM, Green RA, Kaplan KB: APC mutations lead to
cytokinetic failures in vitro and tetraploid genotypes in Min
mice. J Cell Biol 2007, 178:1109-1120.
55. Li Z, Näthke IS: Tumor-associated NH2-terminal fragments are
the most stable part of the adenomatous polyposis coli
protein and can be regulated by interactions with COOHterminal domains. Cancer Res 2005, 65:5195-5204.
56. Green RA, Wollman R, Kaplan KB: APC and EB1 function
together in mitosis to regulate spindle dynamics and
chromosome alignment. Mol Biol Cell 2005, 16:4609-4622.
57. Berrueta L, Tirnauer JS, Schuyler SC, Pellman D, Bierer BE: The
APC-associated protein EB1 associates with components of
the dynactin complex and cytoplasmic dynein intermediate
chain. Curr Biol 1999, 9:425-428.
58. Strickland LI, Wen Y, Gundersen GG, Burgess DR: Interaction
between EB1 and p150glued is required for anaphase astral
microtubule elongation and stimulation of cytokinesis. Curr
Biol 2005, 15:2249-2255.
59. McCartney BM, McEwen DG, Grevengoed E, Maddox P,
Bejsovec A, Peifer M: Drosophila APC2 and Armadillo
participate in tethering mitotic spindles to cortical actin. Nat
Cell Biol 2001, 3:933-938.
60. Amos-Landgraf JM, Kwong LN, Kendziorski CM, Reichelderfer M,
Torrealba J, Weichert J, Haag JD, Chen KS, Waller JL, Gould MN
et al.: A target-selected Apc-mutant rat kindred enhances the
modeling of familial human colon cancer. Proc Natl Acad Sci U
S A 2007, 104:4036-4041.
This paper introduces a novel rat model for Apc-mediated tumourigenesis
that mimics the human disease more accurately.
61. Sansom OJ, Meniel VS, Muncan V, Phesse TJ, Wilkins JA,
Reed KR, Vass JK, Athineos D, Clevers H, Clarke AR: Myc
deletion rescues Apc deficiency in the small intestine. Nature
2007, 446:676-679.
This paper demonstrates that Myc is absolutely required for Apc-deficient
cells to survive in intestinal tissue and form tumours and that de-regulated, nuclear b-catenin is not sufficient.
62. Hart MJ, de los Santos R, Albert I, Rubinfeld B, Polakis P:
Downregulation of ß-catenin by human axin and its
association with the APC tumor suppressor, ß-catenin and
GSK3ß. Curr Biol 1998, 8:573-581.
Current Opinion in Cell Biology 2008, 20:186–193