Review
APC: the plot thickens

https://doi.org/10.1016/S0959-437X(99)00016-7Get rights and content

Abstract

Adenomatous polyposis coli (APC) is an important tumour suppressor in the human colon. It is conserved between human and flies, and promotes, together with Axin and glycogen synthase kinase 3 (GSK3), the degradation of the Wnt-signalling effector β-catenin. Recent experiments have shaped our understanding of how Axin and GSK3 function but the role of APC in this process remains elusive.

Introduction

β-catenin and its Drosophila homolog Armadillo are bi-functional proteins: when associated at the plasma membrane with E-cadherin and α-catenin, they mediate cellular adhesion; as ‘free’ cytoplasmic proteins, they transduce the Wnt signal. Unstimulated cells possess a constitutive activity that keeps the free β-catenin protein unstable. Wnt signaling inhibits this activity, stabilising free β-catenin which subsequently translocates to the nucleus. Here, it binds to the transcription factor TCF (T cell factor) and functions as its co-activator to stimulate the transcription of Wnt target genes (Figure 1).

Recent work has demonstrated that the destabilisation of β-catenin in normal cells is mediated by a multi-protein complex. One component of this complex is glycogen synthase kinase 3 (GSK3) whose chief function during development is mediated by β-catenin/Armadillo, and whose kinase activity is inhibited by Wnt signaling 1, 2. GSK3 phosphorylation of β-catenin earmarks this protein for degradation by the proteasome pathway. Mutation of the GSK3-phosphorylation sites in β-catenin causes it to accumulate to high levels, which can lead to cancer in many tissues [3].

Two further components of the β-catenin-destabilising complex are the tumour suppressor adenomatous polyposis coli (APC) and the recently discovered Axin protein. Mutational loss of APC causes β-catenin to accumulate to high levels in human colon epithelial cells [4], eventually leading to cancer. Axin, the product of the mouse fused locus, was shown to antagonise Wnt signalling [5]. In this review, I focus on recent work that has formed our current understanding of how Axin functions together with GSK3 to promote the destabilisation of β-catenin. APC’s function in this process is still poorly understood, and I shall propose a new idea regarding a possible regulatory role of this tumour suppressor.

Section snippets

β-catenin, an Armadillo repeat protein

β-catenin is a protein composed of three modules (Figure 2a): an amino-terminal domain which contains multiple GSK3 phosphorylation sites needed for its degradation, a central domain consisting of 12 so-called ‘Armadillo repeats’, and a carboxy-terminal domain required for its signalling function. The latter confers transcriptional activation in yeast and in mammalian cells 6, 7 and mediates the signalling function of dorsal β-catenin during axis formation in Xenopus embryos [8].

The

Degradation of β-catenin

Cells that are not stimulated by Wnts rapidly degrade free β-catenin. The serine/threonine kinase GSK3, or its Drosophila homolog Shaggy/Zeste-white 3, play a critical role in this process: GSK3 loss causes free β-catenin to accumulate 28, 29. Furthermore, if one of its GSK3 phosphorylation sites is mutated, β-catenin becomes refractory to degradation 4, 30, 31. GSK3 phosphorylation earmarks β-catenin for degradation by the proteasome pathway 32, 33.

Recent work has shed light on the molecular

Axin, a scaffold protein which facilitates phosphorylation of GSK3 substrates

Evidently, GSK3-phosphorylated β-catenin is earmarked for destruction. Therefore, GSK3 phosphorylation of β-catenin is a critical step which is likely to be under exquisite control. A first indication for this came from the observation that, in vitro, β-catenin is not phosphorylated efficiently by GSK3 [29]. This suggested that another protein might be required for this enzymatic reaction.

A likely candidate for such a protein was identified —namely Axin, the product of the mouse locus fused [5]

APC, a well conserved protein promoting the degradation of β-catenin

As outlined above, Axin is thought to facilitate the phosphorylation of GSK3 substrates in the quaternary complex and to be the main target for Wnt-mediated inhibition of GSK3 activity. However, the quaternary complex also contains APC, which begs the question of what the molecular function of APC might be.

Until recently, only one APC gene was known each in humans and in Drosophila; expression of the latter is largely restricted to neuronal cells [56]. During the past year, a second APC has

A regulatory role of APC

Intriguingly, overexpressed Axin is capable of downregulating β-catenin in APC-mutant colon cancer cells 12••, 13, 48•. In line with this, mutant Axin protein that lacks the APC-binding domain (RGS-less Axin) is equally active 12••, 13, if not more so [48], than full-length Axin in destabilising β-catenin in these cells. These experiments indicated that Axin may be able to downregulate β-catenin without APC under certain circumstances. A caveat is that the cancer cells used contain truncated

A taxi function of APC?

APCs are not only associated with the plasma membrane but are also seen in the cytoplasm and in the nucleus of Drosophila and mammalian cells 17••, 69. It is therefore conceivable that APC shuttles forth and back between these subcellular compartments. I would like to suggest an idea that envisages a function of APC in concentrating β-catenin at the plasma membrane.

In this scenario (Figure 4), APC picks up β-catenin in the cytoplasm (e.g. β-catenin immobilised by a putative anchor, see [26••])

Anomalies

Two recent studies 71, 72 appear to contradict the evidence from mammalian cells and from Drosophila that APC antagonises β-catenin. First, overexpression of human or Xenopus APC in ventral cells of Xenopus embryos induces a secondary axis, mimicking overexpression of β-catenin [71]. This has been taken to mean that APC behaves in this assay like a positive Wnt component such as β-catenin itself but the scenario outlined above offers a different explanation. The levels of endogenous Axin are

Conclusions

Rapid progress has been made recently in our understanding of how Axin and GSK3 earmark β-catenin for degradation and how the doomed β-catenin is recognised by the ubiquitination machinery. The function of APC in this process has been recognised for some time, on the basis of findings in colon cancer cells, and recent loss-of-function experiments in Drosophila have provided strong evidence that APC also promotes destabilisation of β-catenin in normal cells. How APC regulates this process

Acknowledgements

I thank Matthew Freeman, Sean Munro and Rob Arkowitz for discussion and comments on the manuscript. I am particularly grateful to Matthew for earmarking various versions of my model for degradation.

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

References (72)

  • Y Marikawa et al.

    β-TrCP is a negative regulator of Wnt/β-catenin signaling pathway and dorsal axis formation in Xenopus embryos

    Mech Dev

    (1998)
  • K Itoh et al.

    Axis determination in Xenopus involves biochemical interactions of axin, glycogen synthase kinase 3 and β-catenin

    Curr Biol

    (1998)
  • MJ Hart et al.

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

    Curr Biol

    (1998)
  • H Yamamoto et al.

    Phosphorylation of axin, a Wnt signal negative regulator, by glycogen synthase kinase-3β regulates its stability

    J Biol Chem

    (1999)
  • C Sakanaka et al.

    Functional domains of axin. Importance of the C terminus as an oligomerization domain

    J Biol Chem

    (1999)
  • W Hsu et al.

    Identification of a domain of Axin that binds to the serine/threonine protein phosphatase 2A and a self-binding domain

    J Biol Chem

    (1999)
  • X Yu et al.

    Ubiquitous expression of a Drosophila Adenomatous polyposis coli homolog and its localisation in cortical actin caps

    Mech Dev

    (1999)
  • P Polakis

    The adenomatous polyposis coli (APC) tumor suppressor

    Biochim Biophys Acta

    (1997)
  • Y Ahmed et al.

    Regulation of armadillo by a Drosophila APC inhibits neuronal apoptosis during retinal development

    Cell

    (1998)
  • J Hülsken et al.

    Tumor-suppressor gene products in cell contacts: the cadherin-APC-armadillo connection

    Curr Opin Cell Biol

    (1994)
  • CE Rocheleau et al.

    Wnt signaling and an APC-related gene specify endoderm in early C. elegans embryos

    Cell

    (1997)
  • JR Miller et al.

    Signal transduction through β-catenin and specification of cell fate during embryogenesis

    Genes Dev

    (1996)
  • PJ Morin et al.

    Activation of β-catenin-Tcf signaling in colon cancer by mutations in β-catenin or APC

    Science

    (1997)
  • J Hülsken et al.

    E-cadherin and APC compete for the interaction with β-catenin and the cytoskeleton

    J Cell Biol

    (1994)
  • S Orsulic et al.

    An in vivo structure-function study of armadillo, the β-catenin homologue, reveals both separate and overlapping regions of the protein required for cell adhesion and for wingless signaling

    J Cell Biol

    (1996)
  • J Behrens et al.

    Functional interaction of an axin homolog, conductin, with β-catenin, APC, and GSK3β

    Science

    (1998)
  • T Nakamura et al.

    Axin, an inhibitor of the Wnt signalling pathway, interacts with β-catenin, GSK-3β and APC and reduces the β-catenin level

    Genes Cells

    (1998)
  • F Hamada et al.

    Negative regulation of Wingless signaling by D-axin, a Drosophila homolog of axin

    Science

    (1999)
  • J Behrens et al.

    Functional interaction of ß-catenin with the transcription factor LEF-1

    Nature

    (1996)
  • X Yu et al.

    A new Drosophila APC homologue associated with adhesive zones of epithelial cells

    Nat Cell Biol

    (1999)
  • Hamada F, Murata Y, Hishida A, Fujita F, Tomoyasu Y, Nakamura M, Yoyoshima K, Tabata T, Ueno N, Akiyama T:...
  • B Rubinfeld et al.

    Binding of GSK3β to the APC–β-catenin complex and regulation of complex assembly

    Science

    (1996)
  • E Brunner et al.

    pangolin encodes a Lef-1 homologue that acts downstream of Armadillo to transduce the Wingless signal in Drosophila

    Nature

    (1997)
  • S Orsulic et al.

    E-cadherin binding prevents β-catenin nuclear localization and β-catenin/LEF-1-mediated transactivation

    J Cell Sci

    (1999)
  • N Funayama et al.

    Embryonic axis induction by the armadillo repeat domain of β-catenin: evidence for intracellular signaling

    J Cell Biol

    (1995)
  • RT Cox et al.

    Membrane-tethered Drosophila Armadillo cannot transduce Wingless signal on its own

    Development

    (1999)
  • Cited by (86)

    • Capsaicin represses transcriptional activity of β-catenin in human colorectal cancer cells

      2012, Journal of Nutritional Biochemistry
      Citation Excerpt :

      These results suggest that capsaicin suppresses β-catenin expression at both transcriptional and posttranslational levels. It is well established that β-catenin is degraded by the 26S proteasome after the covalent binding with ubiquitin [23]. To confirm that the reductions in β-catenin levels by capsaicin treatment are indeed caused by this mechanism, we tested whether capsaicin treatment results in an increase of ubiquitinated β-catenin levels.

    • Htid-1, the human homolog of the Drosophila melanogaster l(2)tid tumor suppressor, defines a novel physiological role of APC

      2007, Cellular Signalling
      Citation Excerpt :

      The faint bands of both Hsp70 and Hsc70 detected in hTid50/hTid48 and APC Ips (Fig. 4C,D) support this explanation. The detection of β-Catenin and GSK-3β in APC Ips (Fig. 4H,I) is consistent with the published data concerning APC's role in stabilization of its cytosolic pool [27,34–36]. As shown in Fig. 4G Asef [41] has been detected in hTid50/hTid48 IP, but not in APC IP.

    View all citing articles on Scopus
    View full text