Where is APC going?

(JCB 154:1105-1109, 2001)

Yuko Mimori-Kiyosue1,2 and Shoichiro Tsukita2,3

Abstract: APC (adenomatous polyposis coli) protein has been thought to be involved in tumor suppression through the Wnt/b-catenin signaling pathway. However, its connections to the cytoskeleton, especially microtubules, are becoming apparent, and the new functions of APC are attracting increasing interest in understanding the role of APC not only in tumorigenesis, but also in normal physiology.

Defects in the APC (adenomatous polyposis coli) tumor suppressor gene is linked to the development of colorectal cancer (reviewed in Nakamura, 1993; Kinzler and Vogelstein, 1996). Somatic mutations in the APC gene occur in the majority of sporadic colorectal tumors, and germline mutations in APC are responsible for familial adenomatous polyposis, an autosomal dominant inherited disease. The APC protein (APC) is a large multi-domain protein with a molecular mass of 300 kD (Fig.1). APCs modular architecture led to attempts to identify binding partners for each domain, and the discovery that APC binds to b-catenin, a protein that functions in cell adhesion as well as in Wnt-based signal transduction, provided the first significant insight into the biological function of APC in molecular terms (Rubinfeld et al., 1993; Su et al., 1993).
Since then, there have been numerous exciting findings regarding the functional relationship between APC and Wnt signaling (reviewed in Cadigan and Nusse, 1997; Peifer and Polakis, 2000). On the other hand, little attention was paid to the subcellular localization of APC. However, another important functional aspect of APC has been abruptly highlighted; its role in association with cytoskeletons. In these studies, of course, the localization of APC has been examined in detail. In this mini-review, we will overview this newly emerging field in the study of APC, paying special attention to its subcellular distribution.

APC at cell-cell adhesion sites
In various polarized epithelial cells, APC has been found at the sites of cell-cell adhesion. In normal mouse intestinal epithelial cells, immunoelectron microscopy showed APC to be localized in the cytoplasm with a significant concentration along the lateral plasma membrane (Miyashiro et al., 1995). The observation that the COOH terminus of APC directly binds to the PDZ domain of hDLG, a human homologue of the Drosophila discs large (DLG) tumor suppressor protein, also favored the notion that APC is associated with cell-cell adhesion sites and/or lateral membranes (Matsumine et al., 1996), since hDLG is distributed along lateral membranes. Also in several cultured cell lines, APC showed some concentration at cell-cell contact sites (Nathke et al., 1996; Fig.2B).
The occurrence of multiple APC proteins in a single species has been reported recently (reviewed in Dikovskaya et al., 2001); APC and APC2/APCL with a smaller molecular mass in human and mouse, and dAPC and dAPC2/E-APC in Drosophila. Their central portion containing armadillo repeats and the b-catenin binding region is fairly conserved, but their COOH-terminal region diversifies significantly. Consequently, in Drosophila, both dAPC and dAPC2/E-APC lack the PDZ-binding motif at their COOH termini, suggesting that they do not interact with DLG. However, recent studies suggested a functional relationship between Drosophila APC and cell-cell adhesion. In Drosophila, dAPC2/E-APC, which is ubiquitously expressed and abundant in epithelial cells, was found to be concentrated at the apicolateral adhesive zones of epithelial cells (McCartney et al., 1999). This junction-specific concentration required an intact actin cytoskeleton, and depletion or mutation of dAPC2/E-APC in embryos resulted in partial defects in the recruitment of Armadillo, a Drosophila homologue of b-catenin, to the junctions (Yu et al., 1999; Townsley and Bienz, 2000). These observations suggest a role for Drosophila APC in the genesis and maintenance of the integrity of cell-cell junctions.

APC on microtubules
In addition to immunohistochemical analyses, overexpression experiments was also conducted to determine the intracellular localization of APC in several cell lines (Munemitsu et al., 1994; Smith et al., 1994). In these studies, unexpectedly, the exogenously expressed wild-type APC appeared to be distributed along microtubules. In agreement with this, APC was shown to directly bind to microtubules at its COOH-terminal basic region and to stabilize microtubules in vitro (Munemitsu et al., 1994), as well as in vivo (Zumbrunn et al., 2001), similarly to conventional MAPs (microtubule-associated proteins). Subsequently, a high-resolution immunofluorescence study of endogenous APC in cultured MDCK cells was performed (Nathke et al., 1996). In these cells, endogenous APC was localized in clusters of puncta near the ends of microtubules at peripheral membrane sites of migrating edges, and this localization appeared to require intact microtubules. However, as a great deal of attention has been focused on the function of APC that pertains to its ability to regulate b-catenin activity, despite Nathke's beautiful images, the relationship of APC to microtubules has been mostly neglected. However, movies were more influential than still images. Analysis of GFP (green fluorescent protein)-tagged APC (APC-GFP) in living Xenopus A6 epithelial cells uncovered a peculiar dynamic behavior of APC within cells (Mimori-Kiyosue et al., 2000a): APC-GFP moved continuously along a subset of microtubules toward their distal ends in an energy-dependent manner and accumulated as granular aggregates at the ends (Fig.2A). These movies, together with the results of other concurrent genetic and immunofluorescence studies on APC, prompted many APC researchers to turn back to microtubules (reviewed in McCartney and Peifer, 2000).

Microtubule search-and-capture mechanism
Microtubules are structurally very dynamic, and their distal (plus) ends are the primary sites of growth and shortening, exhibiting dynamic instability (reviewed in Desai and Mitchison, 1997). Microtubule organization is highly polar, and microtubule dynamics vary considerably between different regions of the cell and stages of the cell cycle, suggesting that they are spatially and temporally controlled. Through intensive observations of such dynamic behavior of microtubules, Kirschner and Mitchison proposed a search-and-capture mechanism: during interconversion between growth and shortening of their plus ends, microtubules search for the sites (e.g. plasma membranes, chromosomes, organelles etc.) with which they interact to capture, followed by stabilization and reorientation of the microtubule-based cytoskeleton (1986) (Fig.3A). However, at that time, the molecular components involved in this search-and-capture mechanism remained undefined.
In recent years, mainly using GFP technology, several proteins, that are specifically concentrated in transient segments at the growing plus ends of microtubules, have been identified (Table 1). These proteins are thought to be copolymerized into plus ends of microtubules, where they remain for a while, and then dissociate from microtubules, allowing the existence of specialized transient segments at the plus ends of microtubules only in the growth phase (reviewed in Sawin, 2000; Schroer, 2001; Schuyler and Pellman, 2001). These findings naturally led to the hypothesis that these transient segments (and also proteins specifically associated with these segments) play a crucial role in the search-and-capture mechanism of microtubules. Interestingly, from the viewpoint of the APC study, EB1, which was identified as an APC binding protein by yeast two-hybrid screening, was also included in this category of proteins (Su et al., 1995; Mimori-Kiyosue et al., 2000b, reviewed in Tirnauer and Bierer, 2000).
Recent studies in yeast showed that EB1 acts as a crosslinker between microtubule ends and the cell cortex. Bim1p, a budding yeast homologue of EB1, was identified as a tubulin-binding protein whose deletion causes defects in orienting spindles (Schwartz et al., 1997). In the budding yeast, the spindle microtubules search for and capture the tip of daughter buds to align spindles and thereby segregate the nucleus correctly (Fig.3 B). Genetic analyses have revealed that spindle orientation requires several polarity proteins that localize to the tip of buds, including Kar9p which was originally identified in a screen for karyogamy mutants as a protein involved in nuclear migration (Miller and Rose, 1998). Interestingly, Bim1p was found to directly interact with Kar9p and recruit it to microtubules. (Lee et al., 2000; Korinek et al., 2000). Therefore, it is now believed that Bim1p on the plus ends of microtubules captures Kar9p at the tips of buds to assist spindle orientation and faithful cell division. Intriguingly, EB1 is conserved in a wide range of organisms from yeast to human, but no Kar9p homologues have yet been found in other species. This raises the question of the identity of the counterpart of Kar9p in vertebrates. Taking the affinity between EB1 and APC as well as the subcellular localization of APC, it is tempting to speculate that APC is one of the functional counterparts of Kar9p in multicellular organisms. Of course, this speculation should be evaluated experimentally in more detail, since EB1 and APC show no structural similarity.

APC during mitosis
If APC is one of counterparts of Kar9p, it is likely that, also in higher organisms, the APC-EB1-based microtubule-capturing process occurs during cell division (Fig.3C). This led Jan and colleagues (Lu et al., 2001) to test the function of dAPC2/E-APC in epithelial cell division in Drosophila embryos. Drosophila neuroepithelial cells divided in a symmetric manner, but when adherens junctions were destroyed, they changed to divide in an asymmetric manner. Similarly, depletion of dAPC2/E-APC or Drosophila homologue of EB1 (dEB1) by the RNA interference (RNAi) method induced asymmetric cell division. Although the direct binding between dAPC2/E-APC and dEB1 was not detected in vitro, these findings suggested the involvement of dAPC2/E-APC and dEB1 in determination of spindle orientation. Moreover, in dividing neuroblasts, dAPC2/E-APC was shown to be asymmetrically localized at the cortex in a crescent adjacent to one spindle pole (McCartney et al., 1999), suggesting the involvement of dAPC2/E-APC also in asymmetric cell division.
Recently, two independent groups reported that in mitotic cells APC is localized at kinetochores, the microtubule attachment sites of chromosomes, and that APC mutant cells are defective in spindle formation and chromosome segregation (Kaplan et al., 2001; Fodde et al., 2001). Furthermore, Kaplan et al. showed that APC forms a complex with cell-cycle checkpoint proteins, Bub1 and Bub3, at kinetochores, and proposed a model in which APC monitors the accurate attachment of microtubule ends to kinetochores. These findings led to the intriguing hypothesis that APC (and probably EB1) is essential for microtubules to search for and capture the kinetochores during cell division. In the contrast, it was reported that the APC-EB1 interaction is down-regulated in mitotic cells (Askham et al., 2000). Therefore, the above hypothesis should be evaluated experimentally in future studies.

APC during cell migration
The search-and-capture mechanism of microtubules could also work in migrating cells. During migration, microtubules are asymmetrically organized facing their plus ends toward the leading edge of the cell. When the wound healing process was observed using A6 cells expressing GFP-APC, at the front row of the wound GFP-APC began to gradually concentrate at the distal ends of a subset of microtubules, which appeared to grow continuously toward the wounded region (Mimori-Kiyosue et al., 2000a). EB1 appeared to be colocalized with APC only at these ends of microtubules (Mimori-Kiyosue et al., 2000b). Furthermore, APC appeared to associate with microtubules preferentially in migrating epithelial cells, but not in highly polarized cells (Nathke et al., 1996) (Fig.2B). These findings suggested that in migrating cells the APC-EB1 interaction plays some important role in guiding microtubule plus ends to specific cortical sites, as observed in the tips of daughter buds of yeasts.
Recently, the intimate relationship between APC and the actin-based cytoskeleton was also discussed. Asef, APC-stimulated guanine nucleotide exchange factor (GEF), was identified as one of the binding partners for the armadillo repeat region of APC (Kawasaki et al., 2000). APC was shown to enhance the GEF activity of Asef, resulting in the activation of Rac, a small G protein, followed by actin polymerization at the cell periphery, i.e. membrane ruffling, and lamellipodia formation, in MDCK cells. This finding may provide an important clue to understand how APC is involved in the regulation of not only microtubule-based but also actin-based cytoskeletons.

APC in nucleus, apical membranes, ------
We should point out that there has been some controversial reports on the subcellular localization of APC. For example, APC was reported to be localized in the nucleus in several cellular systems (e.g. Neufeld and White, 1997), but this nuclear localization still remains somewhat controversial (Nathke et al., 1996). Furthermore, APC was recently reported to be concentrated at apical plasma membranes in a variety of polarized epithelial cells (Reinacher-Schick and Gumbiner, 2001), but this appears inconsistent with most of the previous observations. Indeed, in highly polarized A6 cells expressing APC-GFP no intense signal was detected from apical membranes (Fig.2B), although exogenously expressed APC-GFP in culture cells does not always mirror the behavior of endogenous protein. In general, we have often encountered difficulty in detection of endogenous APC molecules by immunofluorescence microscopy, partly due to the low expression level of endogenous APC, and partly due to problems in the specificity of commercially available antibodies. After careful consideration, we should discuss where APC will go in the future study.

Where will APC go?
In recent years, based on the subcellular localization and dynamic behavior of APC, several new aspects of APC have emerged. Interestingly, most of these new aspects appear to be related to the cytoskeleton, in particular, the organization of microtubule networks. In this mini-review, we presented an overview of these new findings along with the search-and-capture mechanism of microtubules. This scenario, especially the relationship between APC and Kar9p, should be evaluated experimentally in more detail, but the scenario discussed here may be helpful for researchers to appreciate that the challenge of answering many questions lies ahead before understanding the whole picture of APC functions.
Patients suffered from familial adenomatous polyposis develop hundreds to thousands of polyps in the colon and rectum at an early age, a subset of which invariably progress to malignant cancers if not surgically removed. Polyp formation is initiated by abnormal accumulation of the intestinal epithelium at the crypt-villus boundary, where, in the normal intestine, enterocytes migrate up toward the tips of villi maintaining the integrity of a tight layer of cells with concomitant differentiation. These findings suggested the possible involvement of APC not only in the proliferation and differentiation but also in migration and adhesion of epithelial cells (reviewed in Hansken et al., 1994; Polakis, 1997; Bienz and Clevers, 2000). @Indeed, as discussed above, evidence is now accumulating that APC plays a crucial role in cellular migration and adhesion through its associations with the cytoskeleton. Therefore, further analyses of these newly-identified functions of APC will lead to a better understanding of the molecular mechanism of the APC-based tumorigenesis.
From the viewpoint of cancer research, the idea is also intriguing that APC is directly involved in chromosome segregation as well as in microtubule orientation during mitosis. This is particularly attractive when considering the genetic instability and the loss of epithelial polarity during tumorigenesis. In APC mutant mice, intestinal adenomas are polyclonal during the early stage of polyp formation, and this polyclonality appears to be responsible for tumor progression (Merritt et al., 1997). Actually, human colorectal cancer cells were shown to exhibit a marked defect in chromosome segregation (Lengauer et al., 1997). These observations could be explained by the chromosomal instability induced by miss-searching and/or miss-capturing of kinetochores in dividing cells due to APC mutations. On the other hand, the aberrant orientation of the cell-division plane, which also could be induced by APC mutations, may affect the distribution of cells in the intestine, resulting in the loss of normal monolayer organization. Nevertheless, these ideas are still largely speculative, and thus further experimental support is needed to consolidate this presumptive role of APC in cell division as well as tumorigenesis.
Finally, we should discuss the relationship of the new functions of APC summarized in this review to its classic function, i.e. the destruction of b-catenin. Several lines of evidence suggest that the function of APC at the ends of microtubules is regulated by Wnt/b-catenin signaling. Without Wnt signaling, wild-type b-catenin is rapidly degraded and undetectable in the APC clusters at microtubule ends. However, an exogenously expressed stable b-catenin mutant, N-terminally truncated b-catenin (DN-b-catenin), accumulated in the APC clusters. Moreover, expression of DN-b-catenin impaired the migratory properties and formation of cellular extensions of MDCK cells (Barth et al., 1997; Pollack et al., 1997). These findings led to the speculation that increased stability of b-catenin (and resultant accumulation of b-catenin in the APC clusters) suppresses the APC function at microtubule ends, i.e. the extension of cellular processes through stabilization of microtubules. It has been widely accepted that APC regulates the function of b-catenin, but this speculation implies the reverse, i.e. b-catenin regulates the function of APC.
We now know much about the relationship between APC and the Wnt/b-catenin pathway, but the interactions between APC and the cytoskeleton are becoming apparent. The APC-based connections between the Wnt/b-catenin pathway and cytoskeleton are not yet apparent, but are likely to emerge in the coming years. We are only just beginning to seek the missing pieces linking signal transduction to structure and/or vice versa.

----------------------------------------------
We are grateful to Drs.H.Yamauchi and T.Nagasu (KAN Research Institute) for continuous encouragement. Our thanks are also due to Drs.A.Nagafuchi (Kumamoto Univ.), A.Kikuchi (Hiroshima Univ.) and H.Oda (ERATO) for their critical reading of this manuscript.

Movies showing the dynamic behaviors of APC and EB1 are supplemented in Mimori-Kiyosue et al. (2000a), Mimori-Kiyosue et al. (2000b), and Eccleston (2001).
References
Askham,J.M., P.Moncur, A.F.Markham, and E.E.Morrison. 2000. Regulation and function of the interaction between the APC tumor suppressor protein and EB1. Oncogene 19:1950-1958.
Barth,A.I., A.L.Pollack, Y.Altschuler, K.E.Mostov, and W.J.Nelson. 1997. NH2-terminal deletion of b-catenin results in stable colocalization of mutant b-catenin with adenomatous polyposis coli protein and altered MDCK cell adhesion. J.Cell Biol. 136:693-706.
Bienz,M., and H.Clevers. 2000. Linking colorectal cancer to Wnt signaling. Cell 103:311-320.
Cadigan,K.M., and R.Nusse. 1997. Wnt signaling: a common theme in animal development. Genes Dev. 11:3286-3305.
Desai,A., and T.J.Mitchison. 1997. Microtubule polymerization dynamics. Annu.Rev.Cell Dev.Biol. 13:83-117.
Dikovskaya,D., J.Zumbrunn, G.A.Penman, and I.S.Nathke. 2001. The adenomatous polyposis coli protein:in the limelight out at the edge. Trends in Cell Biology in press.
Eccleston,A. 2001. GFP in Motion 2. Trends in Cell Biology 11:311.
Fodde,R., J.Kuipers, C.Rosenberg, R.Smits, M.Kielman, C.Gaspar, J.H.van Es, C.Breukel, J.Wiegant, R.H.Giles, and H.Clevers. 2001. Mutations in the APC tumour suppressor gene cause chromosomal instability. Nat.Cell Biol. 3:433-438.
Hansken,J., J.Behrens, and W.Birchmeier. 1994. Tumor-suppressor gene products in cell contacts: the cadherin-APC-armadillo connection. Curr.Opin.Cell Biol. 6:711-716.
Kaplan,K.B., A.A.Burds, J.R.Swedlow, S.S.Bekir, P.K.Sorger, and I.S.Nathke. 2001. A role for the Adenomatous Polyposis Coli protein in chromosome segregation. Nat.Cell Biol. 3:429-432.
Kawasaki,Y., T.Senda, T.Ishidate, R.Koyama, T.Morishita, Y.Iwayama, O.Higuchi, and T.Akiyama. 2000. Asef, a link between the tumor suppressor APC and G-protein signaling. Science 289:1194-1197.
Kinzler,K.W., and B.Vogelstein. 1996. Lessons from hereditary colorectal cancer. Cell 87:159-170.
Kirschner,M., and T.Mitchison. 1986. Beyond self-assembly: from microtubules to morphogenesis. Cell 45:329-342.
Korinek,W.S., M.J.Copeland, A.Chaudhuri, and J.Chant. 2000. Molecular linkage underlying microtubule orientation toward cortical sites in yeast. Science 287:2257-2259.
Lee,L., J.S.Tirnauer, J.Li, S.C.Schuyler, J.Y.Liu, and D.Pellman. 2000. Positioning of the mitotic spindle by a cortical-microtubule capture mechanism. Science 287:2260-2262.
Lengauer,C., K.W.Kinzler, and B.Vogelstein. 1997. Genetic instability in colorectal cancers. Nature 386:623-627.
Lu,B., F.Roegiers, L.Y.Jan, and Y.N.Jan. 2001. Adherens junctions inhibit asymmetric division in the Drosophila epithelium. Nature 409:522-525.
Matsumine,A., A.Ogai, T.Senda, N.Okumura, K.Satoh, G.H.Baeg, T.Kawahara, S.Kobayashi, M.Okada, K.Toyoshima, and T.Akiyama. 1996. Binding of APC to the human homolog of the Drosophila discs large tumor suppressor protein. Science 272:1020-1023.
McCartney,B.M., H.A.Dierick, C.Kirkpatrick, M.M.Moline, A.Baas, M.Peifer, and A.Bejsovec. 1999. Drosophila APC2 is a cytoskeletally-associated protein that regulates wingless signaling in the embryonic epidermis. J.Cell Biol. 146:1303-1318.
McCartney,B.M., and M.Peifer. 2000. Teaching tumour suppressors new tricks. Nat.Cell Biol. 2:E58-60.
Merritt,A.J., K.A.Gould, and W.F.Dove. 1997. Polyclonal structure of intestinal adenomas in ApcMin/+ mice with concomitant loss of Apc+ from all tumor lineages. Proc Natl Acad Sci U S A. 94:13927-13931.
Miller,R.K., and M.D.Rose. 1998. Kar9p is a novel cortical protein required for cytoplasmic microtubule orientation in yeast. J.Cell Biol. 140:377-390.
Mimori-Kiyosue,Y., N.Shiina, and S.Tsukita. 2000a. Adenomatous polyposis coli (APC) protein moves along microtubules and concentrates at their growing ends in epithelial cells. J.Cell Biol. 148:505-518.
Mimori-Kiyosue,Y., N.Shiina, and S.Tsukita. 2000b. The dynamic behavior of the APC-binding protein EB1 on the distal ends of microtubules. Curr.Biol. 10:865-868.
Miyashiro,I., T.Senda, A.Matsumine, G.H.Baeg, T.Kuroda, T.Shimano, S.Miura, T.Noda, S.Kobayashi, M.Monden et al. 1995. Subcellular localization of the APC protein: immunoelectron microscopic study of the association of the APC protein with catenin. Oncogene. 11:89-96.
Munemitsu,S., B.Souza, O.Muller, I.Albert, B.Rubinfeld, and P.Polakis. 1994. The APC gene product associates with microtubules in vivo and promotes their assembly in vitro. Cancer Res. 54:3676-3681.
Nakamura,Y. 1993. The role of the adenomatous polyposis coli (APC) gene in human cancers. Adv.Cancer Res. 62:65-87.
Nathke,I.S., C.L.Adams, P.Polakis, J.H.Sellin, and W.J.Nelson. 1996. The adenomatous polyposis coli tumor suppressor protein localizes to plasma membrane sites involved in active cell migration. J.Cell Biol. 134:165-179.
Neufeld,K.L., and R.L.White. 1997. Nuclear and cytoplasmic localizations of the adenomatous polyposis coli protein. Proc.Natl.Acad.Sci.USA. 94:3034-3039.
Peifer,M., and P.Polakis. 2000. Wnt signaling in oncogenesis and embryogenesis--a look outside the nucleus. Science 287:1606-1609.
Polakis,P. 1997. The adenomatous polyposis coli (APC) tumor suppressor. Biochim.Biophys.Acta. 1332:F127-147.
Pollack,A.L., A.I.M.Barth, Y.Altschuler, W.J.Nelson, and K.E.Mostov. 1997. Dynamics of b-catenin interactions with APC protein regulate epithelial tubulogenesis. J.Cell Biol. 137:1651-1662.
Reinacher-Schick,A, B.M.Gumbiner. 2001. Apical membrane localization of the adenomatous polyposis coli tumor suppressor protein and subcellular distribution of the beta-catenin destruction complex in polarized epithelial cells. J.Cell Biol. 152:491-502.
Rubinfeld,B., B.Souza, I.Albert, O.Muller, S.H.Chamberlain, F.R.Masiarz, S.Munemitsu, and P.Polakis. 1993. Association of the APC gene product with b-catenin. Science 262:1731-1734.
Sawin,K.E. 2000. Microtubule dynamics: the view from the tip. Curr.Biol. 10:R860-862.
Schroer,T.A. 2001. Microtubules don and doff their caps: dynamic attachments at plus and minus ends. Curr.Opin.Cell Biol. 13:92-96.
Schuyler,S.C., and D.Pellman. 2001. Microtubule plus-end-tracking proteins: The end is just the beginning. Cell 105:421-424.
Schwartz,K., K.Richards, and D.Botstein. 1997. BIM1 encodes a microtubule-binding protein in yeast. Mol.Biol.Cell 8:2677-2691.
Smith,K.J., D.B.Levy, P.Maupin, T.D.Pollard, B.Vogelstein, and K.W.Kinzler. 1994. Wild-type but not mutant APC associates with the microtubule cytoskeleton. Cancer Res. 54:3672-3675.
Su,L.K., M.Burrell, D.E.Hill, J.Gyuris, R.Brent, R.Wiltshire, J.Trent, B.Vogelstein, and K.W.Kinzler. 1995. APC binds to the novel protein EB1. Cancer Res. 55:2972-2977.
Su,L.K., B.Vogelstein, and K.W.Kinzler. 1993. Association of the APC tumor suppressor protein with catenins. Science 262:1734-1737.
Tirnauer,J.S., and B.E.Bierer. 2000. EB1 proteins regulate microtubule dynamics, cell polarity, and chromosome stability. J.Cell Biol. 149:761-766.
Townsley,F.M., and M.Bienz. 2000. Actin-dependent membrane association of a Drosophila epithelial APC protein and its effect on junctional Armadillo. Curr.Biol. 10:1339-1348.
Yu,X., L.Waltzer, and M.Bienz. 1999. A new Drosophila APC homologue associated with adhesive zones of epithelial cells. Nat.Cell Biol. 1:144-151.
Zumbrunn,J., K.Kinoshita, A.A.Hyman, and I.S.Nathke. 2001. Binding of the adenomatous polyposis coli protein to microtubules increases microtubule stability and is regulated by GSK3 beta phosphorylation. Curr.Biol. 11:44-49.

Table 1

Proteins concentrated at growing microtubule plus ends
in multi-cellular organisms

EB/RP family proteins
EB1
RP1
Cytoplasmic linker proteins and its binding proteins
CLIP -170
CLASPs (CLIP-associating proteins)
Dynein, dynactin complex and their binding proteins
Dynein
p150Glued
Dynamitin/p50
Arp1
LIS-1