Regulation of -catenin phosphorylation and nuclear...

42
1 © 2014. Published by The Company of Biologists Ltd. Journal of Cell Science Accepted manuscript JCS Advance Online Article. Posted on 4 February 2014

Transcript of Regulation of -catenin phosphorylation and nuclear...

1

Regulation of β-catenin phosphorylation and nuclear/cytoplasmic transport by

APC and its cancer-related truncated form.

Authors,

Lili Wang1,2,*, Xiaoyong Liu2, Ekaterina Gusev2, Chuanxin Wang1#, and François Fagotto2, *

Affiliations:

1Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, China, and

2Department of Biology, McGill University, Montreal, Canada

*To whom correspondence should be addressed, François Fagotto, Department of Biology, McGill University 1205 Dr. Penfield Ave., room W5/15 Montreal, QC H3A 1B1, Canada Email, [email protected]

#, Equal last author, Chuanxin Wang, Department of Clinical Laboratory, Qilu Hospital, Shandong University 107 Wenhua Xi Road Jinan, 250012, P.R. China Email, [email protected]

© 2014. Published by The Company of Biologists Ltd.Jo

urna

l of C

ell S

cien

ceA

ccep

ted

man

uscr

ipt

JCS Advance Online Article. Posted on 4 February 2014

2

Abstract

We report the first direct analysis of the endogenous β-catenin phosphorylation activity in colon

cancer SW480 cells. By comparing parental SW480 cells that harbor a typical truncated APC form,

cells expressing full length APC and APC-depleted cells, we provide the formal demonstration that

APC is necessary for β-catenin phosphorylation, both for priming at residue serine 45 and for the

subsequent phosphorylation of residues 33, 37 and 41. Truncated APC still sustains a surprisingly high

phosphorylation activity, which requires β-catenin binding to 20AA repeats for APC, thus providing

biochemical explanation for the precise truncations found in cancer cells. We also discovered that

most of the β-catenin phosphorylation activity is associated with a dense insoluble fraction. We finally

examined the impact of full length and truncated APC on β-catenin nuclear transport. We observed

that β-catenin was transported much faster than previously thought. While this fast translocation was

largely insensitive to the presence of wild type or truncated APC, the two forms appeared to limit the

pool of β-catenin available for transport, which could have an impact on β-catenin nuclear activities in

normal and cancer cells.

Introduction

The Wnt-β-catenin pathway is a major signaling route that controls embryonic patterning and tissue

homeostasis. Its deregulation is involved in many cancers. The pathway is in particular over-activated

in virtually all colon cancer, due to mutations of the adenomatous polyposis coli (APC) tumor

suppressor gene, which could actually represent the initiating event for this type of cancer (Polakis,

2007). The pathway revolves around β-catenin, which, among many other functions, is responsible for

the transduction of Wnt signals into gene regulation through its ability to act as transcriptional

coactivator (Valenta et al, 2012). β-catenin appears to be controlled in the cytoplasm by a complex

built on the scaffold protein Axin. In the absence of Wnt signal, β-catenin is inactivated by the Axin

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

3

complex, also called “β-catenin destruction complex”, Soluble β-catenin is captured by Axin, and

sequentially phosphorylated, first by casein kinase 1 (CK1) of serine residue 45, which serves as

priming for the subsequent action of glycogen synthase 3 (GSK3) on three consecutive residues,

threonine 41, serine 37 and serine 33. N-terminally phosphorylated β-catenin is then ubiquitinated and

rapidly degraded. Upon activation of the pathway by Wnt ligand binding to Frizzled and LRP5/6

receptors, the complex is inhibited by a still poorly understood mechanism (Li et al, 2012; Roberts et

al, 2012; Taelman et al, 2010). This results in the accumulation of soluble β-catenin that can enter the

nucleus, where it interacts with transcription factors of the TCF/LEF1 family to regulate a series of

target genes (Valenta et al, 2012).

Behind this seemingly simple picture of the Wnt pathway, the actual mechanisms that regulate β-

catenin remain highly controversial (Hernandez et al, 2012; Li et al, 2012; Roberts et al, 2012; Taelman

et al, 2010). The role of APC is in particular quite unclear, and the consequences of the mutations

found in cancer cells still ill defined. What appears well established is the fact that the recruitment of

the two kinases, CK1 and GSK3, and their substrate, β-catenin, within a single complex strongly

increases the efficiency of the reaction. It is thus commonly accepted that Axin, CK1 and GSK3

constitute the minimally required “core complex”. Many studies have shown that APC is also essential,

since β-catenin accumulates when APC is mutated or depleted (Munemitsu et al, 1995). That the APC

function is quite proximal to the activity of the Axin complex is strongly suggested by APC’s ability to

associate with both β-catenin and Axin (Fagotto et al, 1999; Hart et al, 1998; Hinoi et al, 2000; Kishida

et al, 1998; Rubinfeld et al, 1993; Su et al, 1993). APC binds directly β-catenin via two different types

of short repeats, called 15 and 20 amino acid repeats. Affinity to the 20AA repeats is strongly increases

by phosphorylation (Ha et al, 2004; Rubinfeld et al, 1996). APC also binds directly to Axin, via short

“SAMP” motifs (Behrens et al, 1998) and indirectly via the Arm repeats (Roberts et al, 2011). APC has

therefore been considered to be a bona fide constituent of the destruction complex (Ha et al, 2004;

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

4

Hinoi et al, 2000; Xing et al, 2003). In one model, Axin and APC are thought to act as coordinate

scaffolds that ensure the specificity of β-catenin phosphorylation and of its regulation by the Wnt

pathway. In vitro experiments using pure recombinant proteins have indeed demonstrated that APC

further increases the efficiency of the Axin-GSK3 complex to phosphorylate β-catenin (Hinoi et al,

2000). The presence of low and high affinity binding sites led to a refined version of this model, which

states that different sites are used depending on the β-catenin levels (Ha et al, 2004). The fact that

phosphorylated 20AA repeats compete with Axin for β-catenin (Ha et al, 2004) suggested a different

model, where APC helped dissociating phosphorylated β-catenin from Axin, creating a catalytic cycle

of binding and release of the substrate (Kimelman & Xu, 2006). Others have suggested that APC acts

either upstream, by gathering or even transporting cytosolic β-catenin to the complex (Bienz, 2002), or

on the contrary downstream of the phosphorylation reactions, recruiting the ubiquitin ligase βTrCP to

the complex (Li et al, 2012; Su et al, 2008). A last interesting variant presented APC functioning both

in β-catenin phosphorylation and its subsequent release from the complex (Roberts et al, 2011).

This uncertainty partly stems from the fact that the central process in this pathway, regulation of β-

catenin phosphorylation, has only been studied in vitro, using purified proteins, or inferred from

observation of steady state levels, rather than by direct measurement of the endogenous kinase

activity. The in vitro data, while demonstrating the role of Axin and APC in making β-catenin

phosphorylation more efficient, also left open a relative wide range of possible reactions. For instance,

GSK3 could phosphorylate β-catenin even in the absence of APC or CK1 (Hinoi et al, 2000), and

even in the absence of Axin, as a matter of fact (Yost et al, 1996), while APC showed some enhancing

effect even in the absence of Axin (Hinoi et al, 2000). Whether APC could also act on the priming

reaction was not tested. A direct transposition of these in vitro data to the in vivo situation is far from

straightforward, since we still know very little about the actual concentrations, activities, associations

and localization of the endogenous components. This raises the question of whether all components

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

5

are required in vivo for the entire process, or whether different partial complexes may be in charge of

partial tasks. Different complexes may also be active under different conditions, for instance low basal

β-catenin levels or high levels during Wnt stimulation (Ha et al, 2004), or even in different cellular

compartments.

Relating the in vitro and in vivo situations would necessarily require investigating specific reactions under

endogenous conditions. Measurement of levels of endogenous phosphorylated β-catenin and

detection of phosphorylated β-catenin at particular cellular locations is clearly not sufficient, and can

be interpreted in opposite ways (e.g. local enrichments could be considered on the contrary as sites of

stabilized β-catenin (Faux et al, 2010). A few studies measured GSK3 activity in the context of the

Wnt pathway, but used substrates irrelevant to the pathway (Stambolic et al, 1996; Taelman et al,

2010). Such measurements almost certainly included activities independent of the Axin-APC complex.

It is even likely that the complex may exclude or at least be poorly accessible to substrates other than

β-catenin. Thus, none of these available data provides adequate information about the actual function

of the pathway. Note also that one still needs to solve the simple question of whether APC is an

intrinsic component of the machinery or merely a modulator. Axin has been indeed considered to be

the limiting factor in the pathway, based on measurements of the relative concentrations in Xenopus

egg extracts (Lee et al, 2003), but see (Tan et al, 2012), and Axin overexpression was found to rescue

lower β-catenin signaling in APC-mutated cancer cells, suggesting that APC may be dispensable when

Axin levels are sufficiently high (Behrens et al, 1998; Cliffe et al, 2003; Faux et al, 2008; Hart et al,

1998); Nakamura et al, 1998).

Considering the many unknowns about APC biology, it comes to no surprise that the precise effect of

the mutations found in colon cancers is similarly unclear and matter of lively debates. The

overwhelming majority of the mutations so far identified in both sporadic (Kinzler & Vogelstein,

1996) and familial colon cancers lead to early termination and thus production of a truncated protein.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

6

Although recessive, these mutations are not random, since many are located in a small region in the

middle of APC coding sequence, indicating that there is a selection in cancer for maintaining the

expression of an intact N-terminus half of the protein (Furuuchi et al, 2000). APC is a very large

(>300kDa) and complex protein, constituted of multiple domains interacting with a variety of cellular

components, and it has been implicated is several different cellular processes, from transcriptional

regulation to mitotic spindle positioning and cell migration (Nathke, 2004). However, the loss-of-

function produced by the deletion of its C-terminus in cancer cells has been definitively linked to the

Wnt pathway (Polakis, 2000). Most truncations remove all but the first of the seven 20AA repeats, as

well as the Axin-binding SAMP repeats (Fig.1A, Kohler et al, 2008). The loss of most of the high

affinity β-catenin binding sites or the inability to bind Axin were thus prime suspects for the abnormal

accumulation of soluble non-phosphorylated β-catenin and for the activation of its transcriptional

targets observed in colon cancer cells. These models have been challenged, however, and different

alternative hypothesis have been proposed, including downstream effects on nuclear localization,

retention or transcription (Bienz, 2002; Krieghoff et al, 2006; Sierra et al, 2006).

As a first step to attempt to clarify some of these issues, we have set a kinase assay to monitor

endogenous activities for β-catenin S45 priming and for S33/S37/T41 phosphorylation. This study

proposes to address the following basic questions, is APC required for β-catenin phosphorylation,

and, if so, for which of the two kinase reactions? What is the effect of APC truncation on β-catenin

phosphorylation?

Another important and still poorly explored question is the subcellular location of the active

complexes responsible for β-catenin phosphorylation. Besides the absence of direct information on

the enzymatic activity, even localization of the components of the complex has not been solidly

established. Detection of endogenous proteins by immunofluorescence has suffered from the lack of

specificity of the antibodies (see e.g. Brocardo et al, 2005), while exogenously expressed constructs

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

7

tend to aggregate (e.g. Fagotto et al, 1999; Faux et al, 2008). In addition, soluble cytosolic components

are known to leak during fixation (Liu & Fagotto, 2011). Studies by cell fractionation, on the other

hand, have been plagued by systematic co-purification of nuclear and plasma membrane insoluble

fractions and by the omission of adequate markers to validate the identity of the fractions (Liu &

Fagotto, 2011). We have recently established a fractionation protocol that cleanly separates the major

cellular components that may be involved in β-catenin regulation (Liu & Fagotto, 2011). Here we have

used this protocol combined to our kinase assay to compare the activity of the various compartments.

The second key process tackled in this study is β-catenin nuclear transport. There have been

conflicting views about a possible role of APC in β-catenin nuclear localization. APC was proposed to

mediate β-catenin export, carrying it via a piggy-back mechanism and that β-catenin nuclear

accumulation in colon cancer cells was due to failure of the truncated APC to fulfill this function

(Bienz, 2002), although the nuclear localization of truncated APC was later contested (Henderson &

Fagotto, 2002) and may be an artifact due to unspecific antibody staining (Brocardo et al, 2005). A

diametrically different mechanism proposed that β-catenin freely shuttles through the nuclear pore

(Fagotto et al, 1998; Koike et al, 2004; Sharma et al, 2012). Kinetic analysis of transport showed that

overexpression of APC or other binding partners such as Axin rather decreased nuclear import

(Krieghoff et al, 2006), supporting the hypothesis that these proteins influenced β-catenin distribution

by sequestration in particular compartments (Krieghoff et al, 2006; Roberts et al, 2011). It remained

however possible that the observed retention was due to the artificial elevated levels of APC and that

endogenous APC may act differently. The effect of truncated APC on β-catenin nuclear translocation

also remained unexplored. We thus included in this study the analysis of β-catenin transport in cells

expressing physiological levels of wild type APC, truncated APC, or cells depleted of APC.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

8

Results

Characterization of components of the Wnt pathway in SW480 and SW480APC cells

In this study, we compared parental SW480 cells with a SW480 cell line stably expressing full length

APC (Faux et al, 2004). We verified that SW480APC cells expressed relatively low levels of full length

APC (Fig. 1B, arrow). Several lower fragments were also detected, including a major band, which,

according to its migration, probably represented the endogenous truncated APC (Fig. 1B, arrowhead).

Note that APC levels are controlled by proteasomal degradation, both wild type and truncated forms

being target for ubiquitination (Choi et al, 2004). Thus one may not necessarily expect identical levels

of the truncated form in the absence of presence of wild type APC. Note also that contribution from a

cleavage product of full the full length protein is also possible, since similar fragments are commonly

observed various cell lines wild type for APC (e.g. Kishida et al, 1998, Liu and Fagotto, unpublished).

The signal was globally decreased in siRNA-transfected cells, demonstrating that all bands were related

to APC (Fig.2C). We also compared the levels of the major components of the Axin complex

(examples in Fig.1B, quantification in supplementary Fig.S1). Axin, GSK3 and CK1 were expressed

roughly at similar levels, with the exception of GSKα, slightly higher in SW480APC. On the contrary,

steady-state levels of β-catenin were lower, consistent with the original report (Faux et al, 2004).

We used our cell fractionation protocol to examine the subcellular distribution of these components.

This protocol yields five fractions (Fig.1B’,B”), cytosol (Cs), nucleosol (Ns), nuclear insoluble fraction

(Ni), membranes (M) and dense insoluble material (X). This latter fraction is mainly composed of

cytoskeleton components, with a minor contribution of nuclear material (Liu & Fagotto, 2011). Most

components of the destruction complex were distributed in similar patterns in parental and APC-

rescued cells (Fig.1B’, quantification in supplementary Fig.1SB). The bulk of Axin, GSK3 and CK1α

were found in the cytosol. On the contrary, CK!ε was strongly enriched in fraction X. Full length APC

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

9

was mostly cytosolic, with a second significant pool in fraction X and low levels in the nucleosol. In

both cell lines, truncated APC was enriched in the cytosol, with smaller pools in nuclear and dense

insoluble fractions (Fig.1B”). As for β-catenin (Fig.1B’), cytosolic levels were high in parental SW80

cells, consistent with previous reports (Munemitsu et al, 1995), but was significantly lower in

SW480APC cells. The dense insoluble fraction contained the second largest pool in both cells lines.

Nucleosolic levels were also slightly lower in SW480APC cells. This characterization whoed that

fraction X constituted the second major subcellular pool for all the components of the destruction

complex. This fraction is generally discarded in cell fractionation experiments, since it has been long

considered as “cell debris”, and has never been analyzed in the context of the Wnt pathway. Note that

Triton X100 was present in the last step of the fractionation, thus fraction X constituted a bona fide

“detergent-insoluble” fraction. We performed APC immunoprecipitation for the Triton-soluble

fraction of the cells (thus fraction X not included). It showed robust co-precipitation of all

components of the destruction complex in both cell lines (supplementary Fig.S2).

β-catenin phosphorylation in SW480 and SW480APC cells

We established a specific in vitro kinase assay to monitor the endogenous activity responsible for β-

catenin N-terminal phosphorylation. Recombinant β-catenin was used as substrate, at the

concentration of 100nM, which corresponds to the estimated cytosolic levels in non-stimulated cells

(Lee et al, 2003). At the dilutions used in this assay, contribution from endogenous β-catenin present

in the cell extracts was negligible (supplementary Fig.S3A). Priming at residue S45 and subsequent

phosphorylation at sites S33/S37/T41 were detected with specific antibodies (pS45 and

pS33/S37/T41) by quantitative immunoblotting (see Materials and Methods). Note that pre-

absorption of the anti-phospho-S33/S37/S41 antibody was essential, because this polyclonal antibody

showed strong cross-reactivity toward non-phosphorylated β-catenin (see Material and Methods).

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

10

Antibody concentrations were optimized to obtain a linear response over the relevant range of signal

intensities.

Comparison of kinase activities in SW480 and SW480APC crude extracts readily yielded an

unambiguous result: the activity was significantly higher in SW480APC cells, both for S45 and for

S33/S37/T41 (Fig.1C). The differences (~ five folds for S45, ~ two folds for S33/S37/T41) were

very reproducible (Fig.1C’), highlighting the robustness of the assay and the consistency of

endogenous activities. Similar difference between the two cell lines were also observed when β-catenin

concentration was raised to 1μM (Supplementary Fig.S3B).

Although not absolutely required for phosphorylation by GSK3 in in vitro experiments, S45 priming is

nevertheless considered essential in vivo. Since full length APC rescue enhanced priming significantly

more than S33/S37/T41 phosphorylation, we wanted to determine whether APC was directly

required for the latter reaction, or if increased S33/S37/T41 phosphorylation was simply a

consequence of accelerated priming. We isolated the S33/S37/T41 phosphorylation step by using a

“constitutively primed” phospho-mimetic β-catenin variant, S45D. We found that the kinase activity

toward S45D β-catenin was also higher in SW480APC cell extracts (Fig.1D). We conclude that full

length APC is required for full activity of both phosphorylation steps. The fact that S33/S37/T41

phosphorylation of wild type and S45D-β-catenin was enhanced to a similar degree indicated that

priming was not limiting in SW480 cells.

The observed two fold increase in overall β-catenin phosphorylation appeared surprisingly modest if

one assumed that APC was absolutely required. Various explanations could account for this rather

mild enhancement: a) Consistent with in vitro experiments, Axin could be sufficient for β-catenin

phosphorylation. APC would then only improve the efficiency of the reaction. b) Axin could be

limiting in these cells, and APC expression could enhance phosphorylation only up to the maximal

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

11

rate allowed by Axin. c) Alternatively, the APC mutation of SW480 cells may not constitute a

complete loss-of-function and the resulting truncated APC may still supply part of APC function in β-

catenin phosphorylation.

To discriminate between these possibilities, we depleted both wild type and truncated APC by RNA

interference (Fig.2A). If truncated APC is not a complete loss-of-function, one would expect that its

depletion would further decrease β-catenin phosphorylation. Otherwise, depletion would have no

effect, and might even increase activity, as the truncated fragment could potentially have an inhibitory

effect on this process. Transfection of APC siRNA led to a ~ 50% depletion of truncated APC in

SW480 cells and ~ 80-90% depletion of full length APC SW480APC cells (Fig. 2C). Both activities

toward S45 and S33/S37/T41 were further reduced, both in SW480 cells and in SW480APC cells. The

decrease was roughly proportional to the reduction in the levels of full length, respectively truncated

APC. Steady-state levels of endogenous β-catenin were also consistently increased (Fig.2C). We

inferred that the presence of APC (or at least of its N-terminal half) was absolutely required for β-

catenin phosphorylation.

We then asked whether Axin was limiting, in which case one would expect that higher levels may

boost β-catenin phosphorylation in SW480APC cells and perhaps even compensate for the absence of

full length APC in SW480 cells. However, mild Axin overexpression (2.3+/-0.4 folds in parental

SW480 cells and 2.5+/-0.9 in SW480APC cells, Fig.2C’) failed to stimulate of β-catenin

phosphorylation (Fig. 2B). On the contrary S45 phosphorylation in SW480 cells was slightly but

reproducibly decreased. We conclude that, contrary to common assumptions, Axin is not limiting, at

least not in SW480 cells and for the specific reaction of β-catenin phosphorylation. These results

further support the notion that APC is crucial and cannot be substituted by increasing Axin levels.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

12

Altogether, these experiments led to two important conclusions: They showed that APC is required

for both phosphorylation steps, and they also demonstrated that truncated APC has still a significant

activity.

Direct binding to APC is required for β-catenin phosphorylation

We verified that the role of APC in β-catenin phosphorylation required direct APC-β-catenin binding.

For this purpose, we used recombinant β-catenin variants with point mutations that specifically

impaired binding to APC. To distinguish between binding to 15 AA and 20 AA repeats (Fig.1A), we

tested three separate mutations (Fig.3), APCΔ15(R386A), defective in binding to 15AA repeats, and

APCΔ20(K345A) and APCΔ20(W383A), both defective in binding to the 20AA repeats (von Kries et

al, 2000). Loss of binding to a specific type of repeat was confirmed by in vitro pull down

(supplementary Fig.S4). These mutant substrates were tested for both S45 and S33/S37/T41

phosphorylation, in SW480 and in APC-rescued cells. In all cases, the activity was lower than for wild

type β-catenin. The difference was relatively mild for the mutant lacking binding to the 15AA repeats,

but quite strong for the two other mutants. Double 345/383 mutation led to a slight but not

statistically significant decrease in the S33/S37/T41 phosphorylation activity compared to the single

mutants. We conclude that both types of interactions are required for full activity, with a stronger

requirement for the 20 AA repeats. These results also highlight the importance of the only 20AA

repeat left in the truncated APC of SW480 cells (see discussion).

β-catenin phosphorylation occurs mainly in an insoluble fraction

We determined the subcellular distribution of β-catenin phosphorylation activity using the above-

mentioned cell fractionation protocol. The distribution of the kinase activity was largely similar in

SW480 and SW480APC cells, and for all three measured activities, i.e. pS45 priming and S33/S37/T41

phosphorylation on wild type β-catenin as well as S33/S37/T41 phosphorylation on constitutively

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

13

primed S45D β-catenin (Fig.4). Quite surprisingly, the dense insoluble fraction “X” was by far the

most active pool accounting for ~ 60-70% of the total cell activity. Comparatively, the cytosol showed

only a modest activity (10-20%), despite the fact that it contained the largest pools of APC, Axin,

CK1α and GSK3 (Fig.1B’, B”). The other significant pool was the nuclear insoluble fraction, which

matched and even surpassed the cytosol in the case of S45D phosphorylation (Fig.4B,D). Nucleosol

and membrane fractions showed low to negligible activity.

Effect of APC and APC truncation on β-catenin nuclear transport

To directly investigate the effect of truncated and wild type APC on β-catenin nuclear transport, we

performed FRAP experiments on SW480 cells transfected with YFP-β-catenin (Fig.5). The import and

export kinetics in parental SW480 cells were roughly similar to those measured in HEK293 and NIH

3T3 cells (Krieghoff et al, 2006; Sharma et al, 2012). However, the use of the spinning confocal

microscope allowed us to obtain information about the initial phase of recovery, which had not been

studied so far. We measured surprisingly fast transport kinetics, both for import and for export (Fig.5

and Table 1). The resulting recovery kinetics clearly fitted a two phase association model, with kinetics

sensibly similar in both directions (Table 1). The first phase of translocation was extremely rapid (K ~

0.1/sec, half-life <10 sec), in fact almost as fast as for GFP, used to monitor free diffusion of a small

protein (Fig.5D,E, supplementary Fig.S5, and Table 1). The second phase was an order of magnitude

slower (K ~ 0.01/sec, half-life > 1 min). Neither import nor export seemed to reach full recovery

after 5 minutes, but approached a plateau around 60-80%. These observations generally suggested the

existence of at least three potential β-catenin pools, one free to diffuse through the nucleopores, the

second subjected to partial retention, and a third pool apparently unavailable for nuclear translocation

on this time scale. Note that β-catenin was transported more efficiently than Cherry-NLS, used as

reporter for classical importin-mediated import (supplementary Fig.S4G,H).

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

14

We also compared transport kinetics of APC-rescued and APC-depleted cells (Fig. 5B-E and Table 1).

For import, the kinetics of the fast phase was sensibly the same under all conditions. We observed

however differences in the contribution of the slow phase (% recovery - % fast phase), which was

larger in APC-rescued cells and smaller in APC-depleted cells. The kinetics of this phase was also

significantly slower in the presence of full length APC, with a half live shifted from ~ 1 min to ~ 5

min. These results indicated that APC acted as a reversible retention component, with the full length

form retaining β-catenin more strongly than the truncated form. As for export, expression of full

length APC had no effect, but depletion again stimulated the process by increasing the fast moving

fraction.

We included in the analysis the effect of inhibition of classical CRM1-mediated export by the drug

Leptomycin B (LMB) (Supplementary Fig.S4A-D). Because nucleocytoplasmic shuttling is a very fast

process, nuclear accumulation of shuttling proteins is expected to be observed within less than one

hours of LMB treatment. In the case of β-catenin, however, a 4-hours treatment had no detectable

effect, neither on import nor on export. In both parental and APC-rescued cells, the recovery curves

perfectly superimposed with those from control cells. In fact, APC distribution under these conditions

was not significantly affected (supplementary Fig.S6). Longer treatments (8hrs) did show however an

effect on import (but not export): Import was altogether increased, with the initial recovery phase

becoming even faster than for APC-depleted cells, approaching the kinetics of free GFP

(supplementary Fig.S5, insert E’).

We also compared the relative nuclear and cytoplasmic steady-state distribution of β-catenin by

measuring the relative fluorescence of YFP-β-catenin in transiently transfected cells. We found that

the nuclear signal was generally close to the cytoplasmic signal (median ~1.2, supplementary Fig. 7A),

although it varied from cell to cell. The ratio was largely similar for parental, APC-rescued and APC-

depleted cells and was not affected by LMB treatment. We also verified that cell to cell variations were

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

15

not related to levels of expression (supplementary Fig.S7B). Note that a very similar ratio was

measured for free GFP, which is considered to freely equilibrate between both compartments. Note

also that even GFP appeared to have an immobile nuclear fraction (suppl. Fig.S5), which probably

account for its nuclear/cytoplasmic ratio being slightly higher than 1.

Discussion

In this study, we have explored systematically three crucial aspects of APC biology. We have

demonstrated the requirement for APC in β-catenin phosphorylation, we have identified that the

activity is largely restricted to an insoluble compartment, and we have formally verified that β-catenin

nuclear transport is independent of APC, which rather acts as its main retention factor. This study has

also confirmed that a typical cancer-related APC truncated form is not a null mutant in terms of Wnt

regulation, but can still promote a significant β-catenin phosphorylation activity. In addition, it still

plays a significant role in β-catenin retention in the context of nuclear transport.

Direct comparison of the function of truncated and full length APC

The APC-rescued SW480 cell line produced by Burgess’ group has provided a powerful tool to

examine APC function in Wnt signaling. Note Faux et al (2004) observed that re-introduction of full

length APC had effects on cell behavior, and in particular on cell adhesion. We have performed a

series of verifications, which did not reveal any overt differences in levels or subcellular distribution of

the components of the Axin complex. We only detected a slight increase in soluble GSK3α, which

does not impact on the interpretation of our results, since the kinase activities are concentrated in the

dense insoluble fraction. Even assuming the existence of other small differences, they would not

account for the dramatic increase in β-catenin phosphorylation measured in APC-rescued cells. A

second issue inherent to cancer cells, and as matter of fact to all immortalized cell lines, is the likely

occurrence of additional unknown mutations. This caveat was circumvented by the comparison with

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

16

siRNA-mediated depleted cells: all our results turned out to be extremely coherent. They can all be

fully explained by the sole contribution of APC.

APC is required for β-catenin phosphorylation

While there is abundant evidence in the literature for a requirement of APC in β-catenin

phosphorylation, this has never been proven in vivo. We provide here the first formal demonstration of

this key point. Our results support those models in which APC is a core component of the Axin

complex (Ha et al, 2004; Hinoi et al, 2000; Roberts et al, 2012; Roberts et al, 2011; Xing et al, 2003).

The intimate involvement of APC in β-catenin phosphorylation is further confirmed by the findings

that APC is required for both S45 priming and subsequent S33/S37/T41 phosphorylation, and that it

requires β-catenin binding directly to APC. While Axin seems to compensate for APC truncations and

rescue “normal” β-catenin in SW480 cells when highly overexpressed (Behrens et al, 1998; Hart et al,

1998), APC seems to be absolutely necessary under more physiological conditions. Note that while

measurement in Xenopus egg extracts suggested that Axin is limiting (Salic et al, 2000; Lee et al, 2003),

Axin and APC are expressed at similar levels in SW480 and SW480APC cells, and Axin is even more

abundant in other mammalian cells (Tan et al, 2012). In Drosophila embryo, β-catenin regulation is

equally sensitive to APC and Axin levels (Roberts et al, 2012; Roberts et al, 2011).

β-catenin phosphorylation occurs in an insoluble fraction

The weak phosphorylation activity of the cytosol was another surprise of our study. The cytosol

seemed indeed to contain an excess of all components required to build active complexes. CK1ε was

the only component absent from this fraction, but CK1α is generally considered to be at least as

effective for β-catenin priming. Dilution is unlikely to explain this low activity, because in our assays

cytosolic fractions were in fact more concentrated than the crude extracts. Furthermore, according to

our immunoprecipitation data (supplementary Fig.S2), all interactions seemed to resist well dilution.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

17

Note also that the conditions for cytosol extraction were milder than those used for

immunoprecipitation (very low digitonin as opposed to high Triton X100). We conclude to the

existence of large cytosolic Axin and APC pools that are poorly or not active at all in β-catenin

phosphorylation. We suggest that cytosol complexes may either participate to a dynamic, perhaps

regulated, equilibrium with fully active insoluble complexes, and/or fulfill other functions, such as

JNK signaling for Axin or cytoskeleton regulation for APC.

Several studies have investigated the possible nature of APC complexes (Mahadevaiyer et al, 2007;

Maher et al, 2009; Penman et al, 2005; Reinacher-Schick & Gumbiner, 2001). A systematic study by

cell fractionation showed that a significant fraction of APC was sedimentable and detergent insoluble

(Reinacher-Schick & Gumbiner, 2001). Other evidence for the association of APC with dense

structures came from the images of APC positive granules in cell protrusions (Mili et al, 2008) or with

the plasma membrane (Reinacher-Schick & Gumbiner, 2001). Junctional localization of APC, Axin

and GSK3 was also reported in SW480 cells (Maher et al, 2009).

Unfortunately, it is difficult to compare our data with any of the previous biochemical investigations,

because they all used so-called “post-nuclear” supernatants of an initial centrifugation. This standard

step of all classical fractionation protocols aims at getting rid of nuclei and unbroken cells. The

content of the discarded pellet, however, contains a large fraction of the cytoskeleton and of the dense

plasma membranes, and significantly overlaps with our fraction X (Liu and Fagotto, 2011). We now

demonstrate that this dense insoluble fraction contains most of the β-catenin phosphorylation activity.

Since this activity fraction was likely missing from all previous analyses (including Bilic et al, 2007; Li

et al, 2012; Taelman et al, 2010), the nature and regulation of the Axin-based β-catenin destruction

complex(es) need to be revisited. Note that a fraction of E-cadherin is also recovered in fraction X,

which is most likely to corresponds to the a detergent-insoluble, cytoskeleton-associated junctional

pool. This probably explains also the relatively high β-catenin levels in this fraction. However, this

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

18

pool is bound to be independent of the Axin-APC complex, since β-catenin interactions with APC

and cadherin are mutually exclusive (Huelsken et al, 1994).

The effect of C-terminal truncations and the relative role of 15 and 20AA repeats

The reason for the strong selection of the cancer-related APC truncations has been abundantly

discussed (e.g. Kohler et al, 2008). The initial theory proposed that loss of the 20AA repeats was

causal to β-catenin stabilization. This view was later challenged and was largely substituted by a model

where the loss of the Axin-binding SAMP motifs had the central role. Recent evidence has re-

questioned this view, showing in particular that APC can interact with Axin independently of SAMP

motifs. It has also become clearer that both the type and number of β-catenin binding repeats are

important in regulation of the Wnt pathway (Roberts et al, 2011). These results have brought back the

original model in the front scene.

In any case, the fact that most truncations left more than half of the APC protein intact indicated that

the distal region of this portion still bore an important function and that there was a strong selection

in cancer cells to preserve it. However, whether this residual function was directly related to β-catenin

phosphorylation has remained an open question. It has been even suggested that the truncated forms

may have acquired some dominant activity. Our data confirm the model of Peifer and colleagues

(Roberts et al, 2011) and unequivocally establish that in terms of β-catenin phosphorylation, APC

truncation behaves as a bona fide loss-of-function, yet not a null but rather a relatively weak allele that

still retains a surprisingly high activity.

Thank to our specific assay, we have been able to further dissect the requirements and confirm/infirm

several previous assumptions: We demonstrate that both types of APC repeats are needed for full

activity. The 20AA repeats are, as predicted, particularly important. This is not only true for cells

expressing full length APC, but even for parental SW480 cells, thus providing a clear explanation for

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

19

the maintenance of one 20AA repeat in truncated APC. On the other hand, we found that binding to

the 15AA, which was thought to have little influence on β-catenin degradation (Roberts et al, 2011;

Roberts et al, 2012), was in fact quite important (Fig.3). The relative contribution of the two types of

repeats correlated quite well with their relative number in full length and truncated APC: the 15AA

repeats played a particularly important role for pS33/S37/T41 phosphorylation in parental SW480

cells, while the 20AA repeats seemed to take care of most of the function for full length APC. These

results do not exclude other specific defects due to APC truncation (we did observe differences in

retention), but in principle the observed decreased rate of β-catenin phosphorylation appears sufficient

to account for its stabilization and over-activation of the pathway.

APC and β-catenin nuclear transport

Although APC (and Axin) was proposed to mediate nuclear export of β-catenin (Bienz, 2002), there is

strong evidence that β-catenin can freely diffuse in and out of the nucleus (Fagotto et al, 1998; Kose et

al, 1997; Wiechens & Fagotto, 2001; Henderson & Fagotto, 2002; Sharma et al, 2012) and that APC

and other partners of β-catenin, all negatively affect β-catenin translocation (Krieghoff et al, 2006).

Our observation of very fast transport kinetics further corroborate the notion of selective diffusion,

while the analysis of transport in APC-depleted cells confirms that APC is not required for

translocation. Note also that the speed of recovery observed in APC-depleted cells, where apparent

retention is very low, approaches the one of freely diffusible GFP, which is quite remarkable, also

considering the differences in size and shape (28kDa and globular for GFP versus rodlike and

>90kDa for β-catenin).

Another definitive argument against a role of APC, or of any other potentially shuttling protein such

as Axin, in β-catenin export is brought by the lack of measurable changes after 4 hrs of LMB-

treatment. In fact, APC does not appear to be a freely shuttling protein, since this 4 hrs treatment is

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

20

not sufficient to cause any significant nuclear accumulation. We had previously obtained similar results

with Axin (Wiechens et al, 2004). It thus seems clear that the ability of Axin and APC to be re-

exported has no short term implications on β-catenin. This property probably serves to maintain on

the long term a proper distribution of these important scaffold proteins in the various cellular

compartments. Obviously complete block of export for longer hours will impact on this distribution,

and eventually also on the pool of β-catenin retained on each side of the nucleus.

In terms of the impact of APC on β-catenin retention, our results perfectly confirm previous results

by Behrens and coworkers (Krieghoff et al, 2006). Our specific contribution is to demonstrate that

APC has measurable effects on β-catenin transport even when expressed at physiological levels. The

fact that retention is significant in APC-expressing cells, and almost nil in the absence of APC suggests

that APC is a major, possibly the main, factor controlling the pool of β-catenin available for transport

in these cells. Consistent with the data of Peifer and colleagues (Roberts et al, 2011; Roberts et al,

2012), truncated APC was still able to retain β-catenin in the cytoplasm, although as argued above, its

high residual activity in β-catenin phosphorylation may have at least as much impact on regulation of

β-catenin signaling in cancer cells. Note that cadherins constitute another component that sequesters

β-catenin at the plasma membrane. Because the association if very strong, we expect that it would

contribute to the immobile fraction in the time scale of our experiments. We do not believe that

cadherins had any significant impact in our FRAP measurements, for the simple reason that we used

single spread cells grown at low density. Under these conditions, cadherin levels are extremely low in

SW480 cells (data not shown). This is consistent with the live images, where GFP-β-catenin was only

weakly and generally not detectable at the membrane, even in cells expressing very low levels of this

construct.

In conclusion, APC truncations are certainly not “null mutants” in term of β-catenin regulation and

still fulfill an unexpectedly large part of APC function, fully consistent with the necessity even for

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

21

cancer cells to maintain β-catenin activity at bay, as proposed in the “just-right” hypothesis (Furuuchi

et al, 2000).

Materials and methods

Cell culture

SW480 cells and SW480APC cells are kind gifts from Antony Burgess (Ludwig Institute, Melbourne,

Australia). Cells were cultured in RPMI 1640 medium, supplemented with 10% fetal bovine serum,

genetecin (1.5 mg/ml) and 1% penicillin/streptomycin.

Antibodies

The antibodies employed in the study were, mouse anti-APC (ALi 12-28, Santa Cruz Biotechnology),

rabbit anti-APC (C-20, Santa Cruz Biotechnology), rabbit anti-APC (M-APC, generous gift of Dr.

Inke Näthke, University of Dundee ( Nathke et al, 1996), affinity purified rabbit anti-Axin (Wiechens

et al, 2004), anti-β-catenin (H102, Santa Cruz Biotechnology), mouse anti-β-catenin (6F9Sigma), rabbit

anti-phospho-β-catenin (Ser33/Ser37/Thr41, Cell Signaling), rabbit anti-phospho-β-catenin (Ser45,

Cell Signaling), mouse anti-GSK3α/β (05-412, Millipore), rabbit anti-casein kinase 1α (sc-28886, Santa

Cruz Biotechnology), goat anti-casein kinase 1ε (sc-6471, Santa Cruz Biotechnology), mouse anti-

casein kinase 1ε (sc-365259, Santa Cruz Biotechnology), mouse anti-GAPDH (6C5, Applied

Biosystems), mouse anti-RanBP3 (BD Biosciences), rabbit anti-LRP6 (C-10, Santa Cruz

Biotechnology), goat anti-γ-Tubulin (sc-7396, Santa Cruz Biotechnology), mouse anti-γ-tubulin

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

22

(ab11316, Abcam), rabbit anti-pericentrin (ab4448, Abcam), rabbit anti-β-actin (ab25894, Abcam), rat

anti-α-actinin (BT-GB-276S, Babraham tech).

Plasmids and recombinant β-catenin construction

eGFP (Clontech), myc-tagged-full length mouse Axin (Zeng et al, 1997), pET-His-β-catenin

(Wiechens & Fagotto, 2001). YFP-βcatenin was constructed by adding the eYFP sequence followed

by a five glycine linker upstream of Xenopus β-catenin in pCS2+MT. CherryNLS was constructed by

adding a classical nuclear localization sequence (KKKRK) to the C terminus of Cherry fluorescent

protein subcloned into the pCS2-vector. Various His-tag-β-catenin and YFP-β-catenin mutants (S45D,

APCΔ15(W386A), APCΔ20(K345A), APCΔ15(W383A) APCΔ20(K345A/W383A) were produced by

site directed mutagenesis based on pCS2-YFP-β-catenin and pET-His-β-catenin using the

QuikChange II XL Site-Directed Mutagenesis Kit, according to the manufacturer’s protocol. All

constructs were confirmed by sequencing. GSTP-APCr15 and r20 were constructed using

oligonucleotides coding for the sequences LDTPINYSLKYSDEQ (1st 15AA repeat of human APC)

and EDTPICFSRCSSLSSLSSAED (1st 20AA repeat). APC-siRNA (sc-29702) and Control siRNA (sc-

37007) were purchased from Santa Cruz Biotechnology. Cells were transfected with plasmids or

siRNAs using Lipofectamine 2000 (Invitrogen) according to the manufacture’s protocol.

Cell homogenization and cell fractionation

For preparation of total homogenates, cells were cultured in 6 cm plastic dishes and harvested by

scraping in 300 μl osmolysis buffer (20mM Hepes-NaOH pH 7.4, 0.2mM EDTA), homogenized with

40 strokes in a tight fitted Douce homogenizer, and equal volume of high Na Buffer (400mM Sucrose,

300mM NaCl, 20mM Hepes-NaOH pH 7.4, 0.2mM EDTA) was added followed by 40 additional

strokes. Our cell fractionation protocol was previously described (Liu & Fagotto, 2011). The

separation yielded five fractions (of the following volumes), cytosol (3 ml), nucleosol (0.5 ml), nuclear

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

23

insoluble (1.35 ml), membranes (1.35 ml) and dense insoluble material (fraction “X”), recovered at the

bottom of the Percoll Gradient and resuspended in 450 μl low Na buffer (150mM NaCl, 10mM

Hepes-NaOH , pH 7.4, 0.1mM EDTA). Note that the Percoll Gradient contained 0.6% Triton-X100

(Liu & Fagotto, 2011).

Western blot

Samples were separated on SDS-PAGE according to the regular protocol, expect for the detection of

APC and Pericentrin which were resolved in a 4% gel without stacking gel. The blots were developed

using a chemiluminescence detection reagent (WBKLS0500, Millipore), and images acquired with a 12

bits digital camera (Alpha Innotech MultiImage system). Data were quantified using the Gene Tools

software (Syngene). Dilution series of were used to verify linearity of the signal. Note that in several

cases a large number of samples had to be blotted simultaneously, which required to run several gels in

parallel. To insure perfectly equal conditions of transfer, incubation with antibodies and development,

two to three gels were transferred on one single nitrocellulose membrane. In all cases presented where

collages are presented, they presented conditions from a single membrane, with identical exposure

time and contrast.

In vitro kinase assay

Substrates were recombinant His-tagged-β-catenin proteins. Proteins were expressed in E. coli BL21

(DE3), purified on Ni-NTA agarose column and exchanged into kinase buffer (150mM NaCl, 20mM

Hepes-NaOH). Reactions were carried out in a total volume of 50μl, containing 100nM recombinant

β-catenin substrates, 1mM ATP, 1mM MgCl2, 10mM Creatine Phosphate and 10U Creatine Kinase,

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

24

and the following amounts of sample, total cell lysates, 20μl; cell fractions, 40μl (undiluted for Cs, Ns,

Ni and Mem fractions, diluted 1:3 in the case of fraction “X”). Volumes were adjusted using kinase

buffer. The reaction was started by adding the samples, was carried on at 37oC, and stopped by

addition of 4x Laemmli sample buffer with 20mM EDTA and boiled immediately for 3 min at 98°C.

Relative levels of phosphorylated S45 and S33/S37/T41 were determined by quantitative immunoblot

using corresponding phospho-specific antibodies. Several commercial anti-phospho-β-catenin

antibodies were tested. Except for the anti-phospho-S45 (Cell Signaling), all other antibodies showed

strong reactivity toward non-phosphorylated β-catenin. This reactivity was eliminated by preabsorbing

anti-phospho-S33/S37/T41 with ~ 50 μg/ml recombinant β-catenin for 60 min before incubation on

the membrane. Band intensities were quantified as above, and relatively activities were calculated after

background subtraction. For samples from crude extracts, as results were expressed as the ratio

between the signal intensity for phosphorylated β-catenin and β-actin used as loading control.

Confocal microscopy and fluorescence recovery after photobleaching

Cells were grown on Fluorodish and transfected with YFP-β catenin, eGFP or CherryNLS. Cells were

maintained in a FCS2 live cell chamber at 37oC, 5% CO2 chamber. Images were acquired using a

Quorum WaveFX spinning disk confocal system (QuorumTechnologies Inc.), with a 40x HCX PL

APO CS, NA= 1.25 oil objective. For photobleaching experiments, samples were photobleached with

a solid state 405nm laser (475mW) using a mosaic digital diaphragm (Andor Technology PLC., Belfast

UK). Either the nucleus or the cytoplasm was bleached for 1 sec at 100% laser power. The samples

were images continuously with a separate 488nm laser line. 5-20 frames of a single z plane were

collected every 200ms before and immediately following bleaching, followed by frames taken at 2-

second intervals. Average nuclear and cytoplasmic intensities were measured using Metamorph or

ImageJ softwares. After background subtraction, the nucleus to cytoplasm (or cytoplasm to nucleus

for export) ratios were calculated. The pre-bleach ratio was set to 100% and ratio in the first post-

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

25

bleach image was set to 0. The recovery curves shown are the averages of at least 8-15 cells from at

least three independent experiments. Curve fitting and statistical calculations were computed using

GraphPad Prism 6.0 and Excel softwares.

Acknowledgements

We thank Drs. Faux and Burgess for generous gift of SW480APC cells, and Laura Canty for providing

the Cherry-NLS construct. We acknowledge the support of the McGill University Biology department

Cell Imaging and Analysis Network (CIAN) for confocal microscopy. L. Wang was recipient of a

Shandong University Joint-Ph.D. training program studentship. This work was supported by a grant

from the Canadian Cancer Research Society to F.F.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

26

References

Behrens, J., Jerchow, B.A., Wurtele, M., Grimm, J., Asbrand, C., Wirtz, R., Kuhl, M., Wedlich,

D., Birchmeier, W. (1998). Functional interaction of an axin homolog, conductin, with beta- catenin,

APC, and GSK3beta. Science 280, 596-599.

Bienz, M. (2002). The subcellular distribution of APC proteins. Nature Rev. Mol. Cell Biol. 3, 328-338

Bilic, J., Huang, Y.L., Davidson, G., Zimmermann, T., Cruciat, C.M., Bienz, M., Niehrs, C.

(2007). Wnt induces LRP6 signalosomes and promotes dishevelled-dependent LRP6 phosphorylation.

Science 316, 1619-1622.

Brocardo, M., Naethke, I.S., Henderson, B.R. (2005) Redefining the subcellular location and

transport of APC, new insights using a panel of antibodies. EMBO Rep 6, 184-190.

Choi , J. Park, S.-Y., Costantini, F., Jho, E.-h., Joo, C.-K. (2004) Adenomatous Polyposis Coli Is

Down-regulated by the Ubiquitin-Proteasome Pathway in a Process Facilitated by Axin. J. Biol. Chem.

279, 49188–49198.

Cliffe, A., Hamada, F., Bienz, M. (2003). A Role of Dishevelled in Relocating Axin to the Plasma

Membrane during Wingless Signaling. Curr. Biol. 13, 960-966.

Fagotto, F., Gluck, U., Gumbiner, B.M. (1998). Nuclear localization signal-independent and

importin/karyopherin- independent nuclear import of beta-catenin. Curr. Biol. 8, 181-190.

Fagotto, F., Jho, E., Zeng, L., Kurth, T., Joos, T., Kaufmann, C., Costantini, F. (1999).

Domains of axin involved in protein-protein interactions, Wnt pathway inhibition, and intracellular

localization. J. Cell Biol. 145, 741-756.

Faux, M.C., Coates, J.L., Catimel, B., Cody, S., Clayton, A.H., Layton, M.J., Burgess, A.W.

(2008). Recruitment of adenomatous polyposis coli and beta-catenin to axin-puncta. Oncogene 27, 5808-

5820.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

27

Faux, M.C., Coates, J.L., Kershaw, N.J., Layton, M.J., Burgess, A.W. (2010). Independent

interactions of phosphorylated beta-catenin with E-cadherin at cell-cell contacts and APC at cell

protrusions. PloS One 5, e14127.

Faux, M.C., Ross, J.L., Meeker, C., Johns, T., Ji, H., Simpson, R.J., Layton, M.J., Burgess,

A.W. (2004). Restoration of full-length adenomatous polyposis coli (APC) protein in a colon cancer

cell line enhances cell adhesion. J. Cell Sci. 117, 427-439.

Furuuchi, K., Tada, M., Yamada, H., Kataoka, A., Furuuchi, N., Hamada, J., Takahashi, M.,

Todo, S., Moriuchi, T. (2000). Somatic mutations of the APC gene in primary breast cancers. Am. J.

Pathol. 156, 1997-2005.

Ha, N.C., Tonozuka, T., Stamos, J.L., Choi, H.J., Weis, W.I. (2004). Mechanism of

phosphorylation-dependent binding of APC to β-catenin and its role in β-catenin degradation. Mol.

Cell 15, 511-521.

Hart, M.J., de los Santos, R., Albert, I.N., Rubinfeld, B., Polakis, P. (1998). Downregulation of

β-catenin by human Axin and its association with the APC tumor suppressor, β-catenin and GSK3β.

Curr. Biol. 8, 573-581.

Henderson, B., Fagotto, F. (2002). The ins and outs of APC and β-catenin nuclear transport.

EMBO Rep. 3, 834-839.

Hernandez, A.R., Klein, A.M., Kirschner, M.W. (2012). Kinetic responses of β-catenin specify the

sites of Wnt control. Science 338, 1337-1340.

Hülsken, J., Birchmeier, W., Behrens, J. (1994) E-cadherin and APC compete for the interaction

with beta-catenin and the cytoskeleton. J. Cell Biol. 127, 2061-2069.

Hinoi, T., Yamamoto, H., Kishida, M., Takada, S., Kishida, S., Kikuchi, A.K. (2000). Complex

formation of adenomatous polyposis coli gene product and axin facilitates glycogen synthase kinase-3

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

28

beta-dependent phosphorylation of β-catenin and down-regulates β-catenin. J Biol Chem 275, 34399-

34406.

Kimelman, D., Xu, W. (2006). β-catenin destruction complex, insights and questions from a

structural perspective. Oncogene 25, 7482-7491.

Kinzler, K.W., Vogelstein, B. (1996). Lessons from hereditary colorectal cancer. Cell 87, 159-170

Kishida, S., Yamamoto, H., Ikeda, S., Kishida, M., Sakamoto, I., Koyama, S., Kikuchi, A.

(1998). Axin, a negative regulator of the Wnt signaling pathway, directly interacts with adenomatous

polyposis coli and regulates the stabilization of β-catenin. J. Biol. Chem. 273, 10823-10826.

Kohler, E.M., Derungs, A., Daum, G., Behrens, J., Schneikert, J. (2008). Functional definition of

the mutation cluster region of adenomatous polyposis coli in colorectal tumours. Hum Mol Genet 17,

1978-1987.

Koike, M., Kose, S., Furuta, M., Taniguchi, N., Yokoya, F., Yoneda, Y., Imamoto, N. (2004).

β-Catenin shows an overlapping sequence requirement but distinct molecular interactions for its

bidirectional passage through nuclear pores. J. Biol. Chem. 279, 34038-34047.

Kose, S., Imamoto, N., Tachibana, T., Shimamoto, T., Yoneda, Y. (1997). Ran-unassisted

nuclear migration of a 97-kD component of nuclear pore-targeting complex. J. Cell Biol. 139, 841-849.

Krieghoff, E., Behrens, J., Mayr, B. (2006). Nucleo-cytoplasmic distribution of Beta-catenin is

regulated by retention. J. Cell Sci. 119, 1453-1463.

Lee, E., Salic, A., Kruger, R., Heinrich, R., Kirschner, M.W. (2003). The roles of APC and Axin

derived from experimental and theoretical analysis of the Wnt pathway. PLoS Biology 1, E10.

Li, V.S., Ng, S.S., Boersema, P.J., Low, T.Y., Karthaus, W.R., Gerlach, J.P., Mohammed, S.,

Heck, A.J., Maurice, M.M., Mahmoudi, T., Clevers, H. (2012). Wnt signaling through inhibition

of β-catenin degradation in an intact Axin1 complex. Cell 149, 1245-1256.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

29

Liu, X., Fagotto, F. (2011). A method to separate nuclear, cytosolic, and membrane-associated

signaling molecules in cultured cells. Sci. Signal. 4, pl2.

Mahadevaiyer, S., Xu, C., Gumbiner, B.M. (2007). Characterization of a 60S complex of the

adenomatous polyposis coli tumor suppressor protein. Biochim. Biophys. Acta 1773, 120-130.

Maher, M.T., Flozak, A.S., Stocker, A.M., Chenn, A., Gottardi, C.J. (2009). Activity of the β-

catenin phosphodestruction complex at cell-cell contacts is enhanced by cadherin-based adhesion.

J.Cell Biol. 186, 219-228.

Mili, S., Moissoglu, K., Macara, I.G. (2008). Genome-wide screen reveals APC-associated RNAs

enriched in cell protrusions. Nature 453, 115-119.

Munemitsu, S., Albert, I., Souza, B., Rubinefeld, B., Polakis, P. (1995). Regulation of

intracellular β-catenin levels by the adenomatous polyposis coli (APC) tumor-suppressor protein. Proc.

Natl. Acad. Sci. USA 92, 3046-3050.

Nakamura, T., Hamada, F., Ishidate, T., Anai, K., Kawahara, K., Toyoshima, K., Akiyama, T.

(1998). Axin, an inhibitor of the Wnt signalling pathway, interacts with β-catenin, GSK-3β and APC

and reduces the β-catenin level. Genes Cells 3, 395-403.

Nathke, I.S. (2004). The adenomatous polyposis coli protein, the Achilles heel of the gut epithelium.

Ann. Rev. Cell Dev. Biol. 20, 337-366.

Nathke, I.S., Adams, C.L., Polakis, P., Sellin, J.H., Nelson, W.J. (1996). The adenomatous

polyposis coli tumor suppressor protein localizes to plasma membrane sites involved in active cell

migration. J. Cell Biol. 134, 165-179.

Penman, G.A., Leung, L., Nathke, I.S. (2005). The adenomatous polyposis coli protein (APC)

exists in two distinct soluble complexes with different functions. J. Cell Sci. 118, 4741-4750.

Polakis, P. (2000). Wnt signaling and cancer. Genes Dev. 14, 1837-1851.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

30

Polakis, P. (2007). The many ways of Wnt in cancer. Curr. Opin. Genet. Dev. 17, 45-51.

Reinacher-Schick, A., Gumbiner, B.M. (2001). Apical membrane localization of the Adenomatous

polyposis coli tumor sppressor protein and subcellular distribution of the beta-catenin destruction

complex in polarized epithelial cells. J. Cell Biol. 152, 491-502.

Roberts, D.M., Pronobis, M.I., Alexandre, K.M., Rogers, G.C., Poulton, J.S., Schneider, D.E.,

Jung, K.C., McKay, D.J., Peifer, M. (2012). Defining components of the ss-catenin destruction

complex and exploring its regulation and mechanisms of action during development. PLoS One 7,

e31284.

Roberts, D.M., Pronobis, M.I., Poulton, J.S., Waldmann, J.D., Stephenson, E.M., Hanna, S.,

Peifer, M. (2011). Deconstructing the β-catenin destruction complex, mechanistic roles for the tumor

suppressor APC in regulating Wnt signaling. Mol. Biol. Cell 22, 1845-1863.

Rubinfeld, B., Albert, I., Porfiri, E., Fiol, C., Munemitsu, S., Polakis, P. (1996). Binding of

GSK3β to the APC-β-catenin complex and regulation of complex assembly. Science 272, 1023-1026.

Rubinfeld, B., Souza, B., Albert, I., Müller, O., Chamberlain, S.H., Masiarz, F.R., Munemitsu,

S., Polakis, P. (1993). Association of the APC gene product with β-catenin. Science 262, 1731-1734.

Sharma, M., Jamieson, C., Johnson, M., Molloy, M.P., Henderson, B.R. (2012). Specific

armadillo repeat sequences facilitate β-catenin nuclear transport in live cells via direct binding to

nucleoporins Nup62, Nup153, and RanBP2/Nup358. J. Biol. Chem. 287, 819-831.

Sierra, J., Yoshida, T., Joazeiro, C.A., Jones, K.A. (2006). The APC tumor suppressor counteracts

β-catenin activation and H3K4 methylation at Wnt target genes. Genes Dev. 20, 586–600.

Stambolic, V., Ruel, L., Woodgett, J.R. (1996). Lithium inhibits glycogen synthase kinase-3 activity

and mimics wingless signalling in intact cells. Curr. Biol. 6, 1664-1668.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

31

Su, L.K., Vogelstein, B., Kinzler, K.W. (1993). Association of the APC tumor suppressor protein

with catenins. Science 262, 1734-1737.

Su, Y., Fu, C., Ishikawa, S., Stella, A., Kojima, M., Shitoh, K., Schreiber, E.M., Day, B.W., Liu,

B. (2008). APC is essential for targeting phosphorylated beta-catenin to the SCFbeta-TrCP ubiquitin

ligase. Mol. Cell 32, 652-661.

Taelman, V.F., Dobrowolski, R., Plouhinec, J.L., Fuentealba, L.C., Vorwald, P.P., Gumper, I.,

Sabatini, D.D., De Robertis, E.M. (2010). Wnt signaling requires sequestration of glycogen synthase

kinase 3 inside multivesicular endosomes. Cell 143, 1136-1148.

Tan, C.W., Gardiner, B.S., Hirokawa, Y., Layton, M.J., Smith, D.W., Burgess, A.W. (2012).

Wnt signalling pathway parameters for mammalian cells. PLoS One 7, e31882.

Valenta, T., Hausmann, G., Basler, K. (2012). The many faces and functions of β-catenin. EMBO

J. 31, 2714-2736.

von Kries, J.P., Winbeck, G., Asbrand, C., Schwarz-Romond, T., Sochnikova, N., Dell'Oro, A.,

Behrens, J., Birchmeier, W. (2000). Hot spots in β-catenin for interactions with LEF-1, conductin

and APC. Nat. Struct. Biol. 7, 800-807.

Wiechens, N., Fagotto, F. (2001). Crm1- and Ran-independent nuclear export of β-catenin. Curr.

Biol. 11, 18-27.

Wiechens, N., Heinle, K., Englmeier, L., Schohl, A., Fagotto, F. (2004). Nucleo-cytoplasmic

shuttling of Axin, a negative regulator of the Wnt-beta-catenin Pathway. J. Biol. Chem. 279, 5263-5267.

Xing, Y., Clements, W.K., Kimelman, D., Xu, W. (2003). Crystal structure of a β-catenin/Axin

complex suggests a mechanism for the β-catenin destruction complex. Genes Dev. 17, 2753-2764.

Yost, C., Torres, M., Miller, J.R., Huang, E., Kimelman, D., Moon, R.T. (1996). The axis-

inducing activity, stability, and subcellular distribution of β-catenin is regulated in Xenopus embryos

by glycogen synthase kinase 3. Genes Dev. 10, 1443-1454.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

32

Zeng, L., Fagotto, F., Zhang, T., Hsu, W., Vasicek, T.J., Perry, W.L.3rd, Lee, J.J., Tilghman,

S.M., Gumbiner, B.M., Costantini, F. (1997). The mouse Fused locus encodes Axin, an inhibitor of

the Wnt signaling pathway that regulates embryonic axis formation. Cell 90, 181-192.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

33

Figure legends

Figure 1

(A) Diagram of human APC and of the truncated form expressed in SW480 cells. The main

domains are represented, including the 15 and 20 amino acid repeats that are responsible for β-catenin

binding, the SAMP motifs that bind Axin, and the B domain (also called β-catenin inhibitory domain),

reported to be required for activity of the destruction complex (Kohler et al, 2008; Roberts et al,

2012). Also included are the confirmed nuclear localization sequences (NLS) and nuclear export

sequences (NES) (Henderson & Fagotto, 2002). MCR, mutation cluster region.

(B-B’’) Expression of the components of the β-catenin destruction complex in parental SW480

cells and in full length APC-rescued cells (SW480APC). (B) Whole extracts. Equal total protein

amounts were loaded. Arrow and arrowhead point respectively to full length (fl) and truncated (tr)

APC. Quantification of relative amounts of full length and truncated APC is presented in

supplementary Figure S1A, and quantification of levels of all the other components in supplementary

Figure S1B. (B’,B”) Subcellular distribution. Cytosolic (Cs), nucleosolic (Ns), nuclear insoluble (Ni),

membrane (M) and dense insoluble (X) fractions were compared between parental (S) and APC-rescue

(A) cells. (B’) APC and β-catenin. Asterisk: non-specific band in parental cells. . (B”) Axin, CK1α and

ε, and GSK. LRP6 and E-cadherin were used as membrane markers, GAPDH and RanBP3 as markers

for cytosol and nucleosol, respectively.See supplementary Fig.S1A for quantification. (B”)

(C,D) Phosphorylation activity toward β-catenin in SW480 and SW480APC cells.

(C,C’) β-catenin phosphorylation was determined in whole extracts using 100nM recombinant β-

catenin as substrate. Levels of phospho-S45 (priming) and phospho-S33/S37/T41 β-catenin were

compared by quantitative immunoblotting using specific antibodies for the corresponding

phosphorylated sites. SW480APC cells showed significantly higher kinase activity than parental cells,

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

34

for both reactions. (C) Western blots from a representative experiment. (C’). Quantification. Relative

intensities were standardized to β-actin input levels. The ratio was arbitrarily set at 1.0 for 15 min

activity in parental SW480 cells. Data are shown as mean intensities ±SD of five independent

experiments. *, **, ***, p<0.05, <0.01, <0.001, respectively, pairwise Student’s t-test.

(D,D’) Recombinant β-catenin with serine 45 mutated to an aspartate (S45D) was used to mimic

constitutive phosphorylation and thus monitor phosphorylation of S33/S37/T41 independently of the

priming step. Phosphorylation was significantly higher in SW480APC cells. (D) Western blots from

representative experiment. (D’) Quantification as for (C).

Figure 2. Effect of APC depletion and Axin overexpression on β-catenin phosphorylation.

(A-A’) APC depletion. Full length and truncated APC forms were depleted in SW480 and

SW480APC cells, respectively, by transfection of siRNA targeting the N-terminal half of the

transcripts. S45 priming and S33/S37/T41 phosphorylation were significantly decreased in both cell

lines. (A) Western blots from representative experiments. (A’) Quantification.

(B-B’) Axin overexpression. SW480 and SW480APC cells transfected with a construct coding for

full length Axin were assayed for β-catenin phosphorylation. Axin overexpression did not increase

phosphorylation. It rather slightly impaired S33/S37/T41 in SW480APC cells. (B) Western blots from

representative experiments. (B’) Quantification. NS, not significant.

(C) Comparison of APC levels in control siRNA and APC siRNA cells. Levels of wild type APC and

truncated APC were decreased respectively to 10-20% and ~50% of normal levels (average of three

experiments). Endogenous β-catenin levels were slightly increased. α-actinin was used as loading

control. (C’) Axin levels in control and YFP-Axin-overexpressing cells. Total Axin levels were

increased 2 to 2.5 folds (average of three experiments). α-actin was used as loading control.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

35

Figure 3. Phosphorylation of β-catenin variants defective in binding to 15AA or 20AA APC

repeats.

Recombinant β-catenin variants were tested that had single amino acid substitutions that impaired

binding to APC, either via 15AA repeats (ΔAPC15(386)) or via the 20AA repeats (ΔAPC20(345),

ΔAPC20(383), and double mutant ΔAPC20(345/383) (von Kries et al, 2000). Compared to wild type

β-catenin, all mutated proteins were significantly less phosphorylated on residues S45 and

S33/S37/T41. β-catenin mutants lacking 20AA binding were the poorest substrates. *, **, ***, p<

0.05, 0.01, and 0.001, respectively, either compared to wild type substrate (just above columns), or to

ΔAPC15(386) (bars).

Figure 4. Subcellular distribution of β-catenin phosphorylation activity.

SW480 (S) and SW480APC (A) cell extracts were fractionated to separate cytosol (Cs), nucleosol (Ns),

nuclear insoluble fraction (Ni), membranes (M), and dense insoluble material (X). The five fractions

were analyzed for S45 and S33/S37/T41 phosphorylation for wild type β-catenin (A) or S45D β-

catenin (B). The results were similar for both cell lines: phosphorylation activity was largely confined

to fraction X. Smaller contributions were observed in Cs and Ni fractions. (C,D) Quantification of

the relative activities. The total kinase activities in SW480 and in SW480APC cells were calculated as

the sum of the activities in the five cell compartments, taking into account the volume of each

fraction. These values were then used to calculate the % contribution from each fraction.

Quantification was performed using the 7.5 min time points, which were most consistent. Although

the kinase activities in fractionated samples tended to be unstable after longer incubations, and thus

less reliable, the general pattern at 15 min was similar.

Figure 5. β-catenin nuclear transport in parental, APC-rescued, and APC-depleted SW480

cells.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

36

SW480 cells, SW480APC rescued cells, and SW80 cells transfected with siAPC were transiently

transfected with YFP-β-catenin. YFP-β-catenin nuclear transport was analyzed by fluorescence

recovery after photobleaching (FRAP). Fluorescence recovery was monitored for 300 sec, and

quantified by plotting the nuclear to cytoplasmic ratio for import, and cytoplasmic to nuclear for

export, setting the pre-bleach fluorescent intensity values to 100% and the post-bleach value to 0%.

(A) Examples of nuclear and cytoplasm FRAP. (B-D) Graph showing compiled data (at least 10 cells

from 3-5 independent experiments).

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

SW480 Cs Ns Ni M X

SW480 APC Cs Ns Ni M X

Figure 1 Wang et al

Dimerization Arm

domain 15 AA repeats 20 AA repeats

SAMP B domain

Basic domain

EB1 binding

NLS NES MCR Truncated APC in SW480 cells

Full length human APC

B

APC β-cat

CK1α

β-actin

Axin

GSK3 β α

SW480 SW480

APC

D’

0

1

2

3

4

0 15 300

5

10

15

20

0 15 30

0

1

2

3

4

0 15 30

phospho S33/S37/T41

phospho S45

β-actin

SW480 SW480APC

0 15 30 0 15 30

wt β-catenin

S45D β-catenin

phospho S33/S37/T41

β-actin

min

SW480 cell

SW480APC cell

Rela

tive

ratio

Rela

tive

ratio

Re

lativ

e ra

tio

phospho S33/S37/T41 phospho S45

phospho S33/S37/T41

min min

min

A

B’

C

C’

D

*

** ***

***

** **

GSK3 β α

CK1ε

LRP6

Axin

CK1α

RanBP3

GAPDH

E-cadh

SW480 SW480

APC

SW A SW A SW A SW A SW A Cs Ns Ni M X B”

tr

fl

*

β-cat

APC tr

fl

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

SW480 cells

A siCTRL siAPC

0 15 30 0 15 30

phospho S33/S37/T41

β-actin

phospho S45

phospho S33/S37/T41

β-actin

phospho S45

Figure 2 Wang et al

siCTRL siAPC 0 15 30 0 15 30

SW480APC cells

phospho S33/S37/T41

β-actin

phospho S45

vector Axin

0 15 30 0 15 30

SW480 cells

B

phospho S33/S37/T41

β-actin

phospho S45

vector Axin

0 15 30 0 15 30

SW480APC cells

Axin

β-actin

C’ C

A’

01234

0 15 3001234

0 15 300

1

2

0 15 300

1

2

0 15 30

siAPC siCTRL

Rela

tive

ratio

pS33/S37/T41 pS45

min min Re

lativ

e ra

tio

pS33/S37/T41 pS45

min min

B’

012345

0 15 300

1

2

3

0 15 300

1

2

3

0 15 300

1

2

3

0 15 30

Rela

tive

ratio

pS33/S37/T41 pS45

min min

Rela

tive

ratio

pS33/S37/T41 pS45

min min Axin vector

siAPC siCTRL

Axin vector

SW480 cells SW480APC cells siCTRL siAPC siCTRL siAPC

SW480 SW480APC

YFP-Axin

vct Axin vct Axin

min min

min min

**

* ** * *

*** *** ***

NS

NS

NS

NS NS

NS

NS NS

β-catenin β-catenin

α-actinin

siCTRL siAPC siCTRL siAPC

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

0

0.2

0.4

0.6

0.8

1

Rela

tive

ratio

0

0.2

0.4

0.6

0.8

1

Rela

tive

ratio

SW480 cells

WT APCΔ15(386) 0 15 30 0 15 30

APCΔ20 (383) 0 15 30 0 15 30

APCΔ20(345) APCΔ20 (345/383) 0 15 30

phospho S45

β-actin

phospho S33/S37/T41

phospho S45

β-actin

phospho S33/S37/T41

SW480APC cells

SW480 cells

phospho S45 phospho S33/S37/T41

0

0.2

0.4

0.6

0.8

1

Rela

tive

ratio

0

0.2

0.4

0.6

0.8

1

Rela

tive

ratio

phospho S45 phospho S33/S37/T41

* *** *** ***

p=0.06

* * *

* ** *** *** **

*** ** ***

* **

** *

Figure 3 Wang et al

min

SW480APC cells

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

Figure 4 Wang et al

pSer33/Ser37/Thr41

A

B

0.0

0.2

0.4

0.6

0.8

1.0

SW APC SW APC SW APC SW APC SW APC

Cs Ns Ni Mem X

0.0

0.2

0.4

0.6

0.8

1.0

SW APC SW APC SW APC SW APC SW APC

Ns Cs Ni Mem X

S A S A S A S A S A Cs Ns Ni M X

0 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 0

½

pSer45

C

pSer33/Ser37/Thr41

S A S A S A S A S A Cs Ns Ni M X

0 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 7.5 15 0

wt β-cat

wt β-cat

S45D β-cat

substrate: wt β-cat

pSer45

substrate: S45D β-cat pSer33/Ser37/Thr41

D

Frac

tion

of to

tal c

ell a

ctiv

ity

0.0

0.2

0.4

0.6

0.8

1.0

SW APC SW APC SW APC SW APC SW APC

Cs Ns Ni Mem X

pSer33/Ser37/Thr41

Frac

tion

of to

tal c

ell a

ctiv

ity

Frac

tion

of to

tal c

ell a

ctiv

ity

min

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

Figure 5 Wang et al

0

20

40

60

80

100

0 100 200 300

SW480SW480 siRNAGFP

0

20

40

60

80

100

0 100 200 300

SW480SWAPC

import

0

20

40

60

80

100

0 100 200 300

SW480SWAPC

export

0

20

40

60

80

100

0 100 200 300

SW480SW480 siRNAGFP

Pre-bleach

Post-bleach 20s 40s 80s 160s

SW480

siRNA

SW480APC

SW480APC

import

export

A

B C

D E Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t

Table 1 Wang et al

IMPORT SW480 siRNA APC

Single curves Whole set Single curves Whole set Single curves Whole set

aver SD total SE aver SD Ttest (toSW) total SE aver SD Ttest

(toSW) total SE

Kfast 0.12 +/-0.02 0.09 +/-0.08 0.15 +/-0.01 0.001 0.14 +/-0.08 0.16 +/-0.04 0.003 0.18 +/-0.13

Kslow 0.006 +/-0.002 0.010 +/-0.010 0.005 +/-0.002 0.07 0.006 +/-0.010 0.003 +/-0.002 0.0004 0.002 +/-0.006

Plateau (% rec)

83 +/-27 58 +/-17 85 +/-19 0.41 94 +/-60 97 +/-30 0.15 104 +/-170

%Fast 28 +/-18 29 +/-14 49 +/-25 0.01 39 +/-17 13 +/-14 0.025 14 +/-20

EXPORT SW480 siRNA APC

Single curves Whole set Single curves Whole set Single curves Whole set

aver SD total SE aver SD Ttest (toSW) total SE aver SD Ttest

(toSW) total SE

Kfast 0.16 +/-0.05 0.15 +/-0.10 0.12 +/-0.05 0.15 0.09 +/-0.10 0.16 +/-0.04 0.5 0.18 +/-0.14

Kslow 0.005 +/-0.003 0.006 +/-0.007 0.009 +/-0.008 0.15 0.004 +/-0.011 0.008 +/-0.006 0.16 0.009 +/-0.005

Plateau (% rec)

53 +/-19 52 +/-30 76 +/-25 0.04 122 +/-187 60 +/-29 0.3 52 +/-12

%Fast 36 +/-23 31 +/-13 35 +/-23 0.5 20 +/-20 30 +/-22 0.3 25 +/-6

Summary table of the main parameters for nuclear import and export obtained from FRAP analysis. Recovery curves (nuclear to cytoplasmic ration for import and cytoplasmic to nuclear ratio for export, see Fig. 7) were fitted with a two phase association algorithm. The table presents, for each condition, average values calculated from fitting of each individual cell measurement, as well as the fitting of compiled data from the whole set of experiments. Both methods gave very similar values. Kfast and Kslow represent kinetics constants for the two phases. Plateau is given as percentage of the pre-bleach value, and corresponds to the “mobile” faction. %Fast indicates the relative contribution of the fast phase to the overall curve.

Jour

nal o

f Cel

l Sci

ence

Acc

epte

d m

anus

crip

t