Tumor suppressor proteins protect cells and tissues from malignant transformation. Among their diverse actions, many of these proteins interact with the microtubule cytoskeleton. This review focuses on the interactions of several tumor suppressors with microtubules and speculates on how disruption of microtubule-dependent processes may contribute to cancer development and spread. We conclude that several tumor suppressors stabilize microtubules and organize microtubule arrays, functions that are likely to be important in preventing tumorigenesis. How tumor suppressors link microtubule stability with cell fate, and how their mutation affects the response of cancer cells to anti-microtubule chemotherapy drugs, remains unclear; these should prove fertile areas for future research.

Microtubules exist in all eukaryotic cells. They form by the polymerization of α/β tubulin dimers into protofilaments, which assemble into hollow tubules. These polymers have the length and tensile strength to act as scaffolds for moving intracellular cargoes across large distances and for controlling cellular architecture. They are highly dynamic, undergoing length and organization changes on time scales ranging from seconds to hours. Most microtubule length changes are accomplished by the addition and removal of tubulin dimers from the end of the tubule termed the plus end. The transitions between polymerization (lengthening) and depolymerization (shortening), referred to as dynamic instability, occur randomly, but they can be altered in response to temporal and spatial cues (Desai and Mitchison, 1997). These alterations are typically mediated by microtubule-associated proteins (MAPs). In a further layer of regulation, tubulin and MAPs are both subject to post-translational modifications (Drewes et al., 1998; Hammond et al., 2008; Howard and Hyman, 2003).

The placement of microtubule minus ends, which influences the organization of the array, is controlled by the centrosome-based microtubule-organizing center (MTOC). MTOCs are rich in the microtubule-nucleating protein γ-tubulin and are surrounded by pericentriolar material, which captures existing microtubule minus ends. The layout of microtubule arrays differs according to cell type and behavior (Fig. 1). Polarized epithelial cells – from which most cancers develop – place their centrosome just below the apical cell surface and extend a diffuse linear microtubule array towards the basal cell surface; apical and basal webs of microtubules are also present (Musch, 2004). In most epithelial cells, centrosomes also nucleate a microtubule-based cilium that protrudes upward from the apical cell surface to sense extracellular flow. Fibroblastic cells, by contrast, anchor their MTOC near the nucleus and extend a radial microtubule array outward to the cell periphery.

Normally, cells have one MTOC that is duplicated early in mitosis. Paired MTOCs migrate to opposite sides of the nuclear envelope to form the two poles of the mitotic spindle. Once the nuclear envelope breaks down, a burst of microtubule polymerization creates the bipolar spindle. Microtubule plus ends then form an elaborate set of contacts with the chromosomes to promote their segregation.

Microtubule plus ends make connections with organelles and other intracellular structures. Proteins that recognize microtubule plus ends [plus-end tracking proteins (+TIPs)] can mediate stable or transient linkages of microtubule plus ends to vesicles, chromosomes and the cell cortex. These connections are vital for vesicle trafficking, cell polarization and migration, chromosome segregation, and spindle orientation within the cell.

Cells alter the spatial organization of their microtubule array in response to internal and external cues. Even subtle disruptions in microtubule length and organization can have profound effects on the cell and may promote cancer development (Mitchison, 1986). Several tumor suppressor proteins stabilize microtubules and control microtubule-dependent processes. Inactivation of these tumor suppressors impairs epithelial polarization and cell division through effects on microtubules, producing characteristics common to many cancers.

An emerging view of cancer (Fig. 2) suggests that normal cells need only to acquire a select set of ‘capabilities’ to become malignant and escape from their tissue of origin. These capabilities include self-stimulation, evasion of restrictive signals, immortalization, angiogenesis and metastasis (Hahn and Weinberg, 2002; Hanahan and Weinberg, 2000). Many of these abilities are acquired by genetic alterations; thus, genomic instability accelerates the acquisition of all the other capabilities (Hahn and Weinberg, 2002; Hanahan and Weinberg, 2000). Changes in microtubule function could account for several of these capabilities, suggesting a common mechanism for tumor suppressors that regulate microtubules in promoting cancer development.

Fig. 1.

Microtubule organization in epithelial cells versus fibroblastic cells. (A) Epithelial cells. The microtubule-organizing center (MTOC) is apically located during interphase. Microtubules run apico-basally and form apical and basal webs. During cell division, spindle poles extend astral microtubules to the adherens junctions to orient the spindle. (B) Migrating fibroblastic cell. The nucleus moves behind the MTOC, which extends a radial microtubule array. Microtubules extending towards the leading edge in the direction of cell migration (arrow) are selectively stabilized. Conversion from an epithelial phenotype to a more fibroblastic behavior is a component of the epithelial-to-mesenchymal transition that characterizes tumor metastasis, and misorientation of the mitotic spindle can alter the balance between stem cell replenishment and stem cell expansion.

Fig. 1.

Microtubule organization in epithelial cells versus fibroblastic cells. (A) Epithelial cells. The microtubule-organizing center (MTOC) is apically located during interphase. Microtubules run apico-basally and form apical and basal webs. During cell division, spindle poles extend astral microtubules to the adherens junctions to orient the spindle. (B) Migrating fibroblastic cell. The nucleus moves behind the MTOC, which extends a radial microtubule array. Microtubules extending towards the leading edge in the direction of cell migration (arrow) are selectively stabilized. Conversion from an epithelial phenotype to a more fibroblastic behavior is a component of the epithelial-to-mesenchymal transition that characterizes tumor metastasis, and misorientation of the mitotic spindle can alter the balance between stem cell replenishment and stem cell expansion.

Close modal

Inactivation of some tumor suppressors destabilizes microtubules

As summarized in Table 1, five tumor suppressors [adenomatous polyposis coli (APC), Ras association domain family 1A (RASSF1A), von Hippel-Lindau (VHL), E-cadherin and merlin] stabilize microtubules in cell-based assays, and their inactivation destabilizes microtubules. An exception is liver kinase B1 (LKB1), which appears to destabilize microtubules in interphase cells and stabilize them in mitotic cells, although the number of studies looking at these roles is small. For most of these proteins, very few biochemical studies are available to guide our understanding of whether their microtubule interactions are direct or indirect. Regardless of the mechanism of microtubule binding, it is likely that most of these proteins act as weak microtubule stabilizers, and that loss of their function reduces microtubule stability to a degree that influences cellular processes but does not kill the cell.

Fig. 2.

Mechanisms by which tumor suppressor mutation-associated microtubule defects could contribute to tumor capabilities. Symbols and associated tumor cell capabilities are adapted from the Hanahan and Weinberg hallmarks wheel (with permission from Elsevier) (Hanahan and Weinberg, 2000), with the addition of a separate symbol for genomic instability. Where applicable, the roles microtubules play in preventing development of these capabilities are listed.

Fig. 2.

Mechanisms by which tumor suppressor mutation-associated microtubule defects could contribute to tumor capabilities. Symbols and associated tumor cell capabilities are adapted from the Hanahan and Weinberg hallmarks wheel (with permission from Elsevier) (Hanahan and Weinberg, 2000), with the addition of a separate symbol for genomic instability. Where applicable, the roles microtubules play in preventing development of these capabilities are listed.

Close modal

Microtubule destabilization may increase cell growth and survival signaling

Two major features of epithelial cell microtubules are their apico-basal polarization and their high degree of stability. Loss of these features may have implications for the trafficking of proteins that affect cell growth and survival. For example, receptor-mediated growth factor signaling may require an array of long, stable microtubules to traffic the receptor from the cell surface to the nucleus. In addition to serving as tracks for transport, microtubules also sequester some of the signaling proteins involved in pro- and anti-growth signaling (Hahn and Weinberg, 2002; Hanahan and Weinberg, 2000). Examples of positive effects on growth signaling include activation of the epidermal growth factor receptor (EGFR) and the estrogen receptor (ER) upon microtubule destabilization (Manavathi et al., 2006). Alternatively, if the cell uses microtubules to sequester signaling proteins, microtubule destabilization could increase the nuclear translocation of these proteins, amplifying signaling cascades that increase cell proliferation (Massague and Weinberg, 1992).

Microtubules also sequester or scaffold some of the proteins involved in apoptosis. Microtubules bind to both pro- and anti-apoptotic regulators, further complicating the analysis of the net effect of microtubule destabilization on cell fate (Manavathi et al., 2006). The pro-survival protein survivin, which is upregulated in many cancers, is an example of an apoptotic regulator whose activity may be affected by changes in microtubule stability (Manavathi et al., 2006). The release of pro-apoptotic proteins into the cytoplasm could tip the balance between pro- and anti-apoptotic signaling, affecting cell fate decisions. Sorting out how microtubule destabilization contributes to apoptosis could be helpful in understanding tumor suppressor mechanisms.

Changes in microtubule organization and stability may contribute to loss of polarity and other epithelial-to-mesenchymal transition-like changes

Most tumors arise from epithelial cells, which form a cohesive sheet that lines the tissue in which they reside. During tumor progression, these cells often lose their epithelial characteristics, including the active maintenance of cell-cell junctions, apico-basal polarization with directional protein sorting, and the capacity for sheet migration that preserves cell-cell and cell-matrix interactions. The epithelial cells detach from the extracellular matrix and from each other, acquire front-back polarity, and use fibroblastic-type migration to egress from the tissue of origin. This constellation of changes has been referred to as an epithelial-to-mesenchymal transition (EMT), or EMT-like phenomenon, after the developmental process (Etienne-Manneville, 2008; Klymkowsky and Savagner, 2009; Polyak and Weinberg, 2009). The EMT is seen in many tumor types, especially at the edges of the tumor, and it is thought to be instrumental in tumor metastasis. Recent evidence has linked the EMT to the acquisition of stem cell behavior (Mani et al., 2008). Indeed, genetic changes that promote such an EMT-like conversion have been shown to facilitate tumorigenesis (Bilder et al., 2000).

Table 1.

Familial syndromes associated with microtubule-interacting tumor suppressors

Familial syndromes associated with microtubule-interacting tumor suppressors
Familial syndromes associated with microtubule-interacting tumor suppressors

EMT-like processes are associated with dramatic reorganizations of the epithelial microtubule array. The linear, highly stable apicobasal array converts to a fibroblastic pattern, collecting minus ends at an MTOC near the nucleus and increasing plus-end dynamic instability. This facilitates fibroblast-type migration, with extension of a leading edge and retraction of the rear of the cell (Wen et al., 2004). Microtubules are required for both maintaining epithelial polarity and generating some of the complex EMT-associated phenotypes (Dugina et al., 1995; Ivanov et al., 2006; Ligon and Holzbaur, 2007; Meng et al., 2008; Shaw et al., 2007; Waterman-Storer et al., 2000; Yap et al., 1995; Yu et al., 2003). Thus, it is possible that changes in microtubule stability and organization that are induced by tumor suppressor mutations could promote EMT-like phenomena that are advantageous to tumor formation or metastasis.

Another way in which microtubule destabilization may contribute to an EMT is by weakening cell-cell junctions that are needed for maintaining epithelial polarity. Interestingly, several microtubule-interacting tumor suppressors localize to cell-cell junctions. E-cadherin forms a crucial homotypic adhesion molecule for these junctions (van Roy and Berx, 2008); APC, LKB1 and merlin localize to junctions; and RASSF1A and VHL have both been proposed to play a role in junction formation, the latter through controlling levels of E-cadherin and tight junction components (Calzada et al., 2006; Dallol et al., 2005; Evans et al., 2007; Harten et al., 2009). Microtubule minus and plus ends connect to cell-cell junctions to deliver junctional components and to transmit polarity information to the cell; loss of these connections has been implicated in junction disassembly (Ligon and Holzbaur, 2007; Shaw et al., 2007; Waterman-Storer et al., 2000; Yap et al., 1995). Microtubule-interacting tumor suppressors are all potential candidates to link microtubules to these junctions, and their inactivation could disrupt epithelial polarity, reduce epithelial barrier function, impair ciliogenesis and alter spindle orientation (Amin et al., 2009; Flaiz et al., 2008; Lallemand et al., 2003; Shibata et al., 1994; Yu et al., 1999).

Microtubule destabilization could also contribute to an EMT-like process by disconnecting microtubules from actin. All of the microtubule-interacting tumor suppressors discussed in this review also bind to actin or an actin regulatory protein (Dallol et al., 2005; Kamada et al., 2001; McClatchey and Fehon, 2009; Nathke, 2005; Perez-Moreno et al., 2003; Tsukita et al., 1992; Zhang et al., 2008). The role of microtubule-actin linkages in cancer protection is unknown, but interactions between the two polymer systems are known to facilitate epithelial polarization and cell migration, as well as mitotic spindle orientation.

Small changes in microtubule regulation may impair chromosome segregation and induce aneuploidy

Probably the most obvious consequence of microtubule destabilization is its effect on mitotic spindle function. Formation of the spindle requires near-complete depolymerization of the microtubule array, followed by a burst of new polymerization. An elaborate set of microtubule plus-end interactions with the chromosomes then occurs. Mitotic spindle defects lead to chromosomal instability (CIN) and aneuploidy, which is associated with many cancer types (Pellman, 2007). Mitotic abnormalities include spindle multipolarity, chromosome misattachments and cytokinesis defects (Fig. 3).

Multipolar spindles form by centrosome overduplication or inheritance of additional centrosomes from a previous cell cycle. Some cells correct spindle multipolarity by clustering centrosomes into a single spindle pole or by expelling them from the cell; some cells with multipolar spindles fail to complete mitosis (Acilan and Saunders, 2008). For cells that do not correct spindle multipolarity, progression through mitosis produces aneuploid daughter cells (Zyss and Gergely, 2009). Multipolar spindles have been found in many tumor types as well as in early carcinoma in situ lesions, suggesting a possible contributory role to the development of invasive cancer (Lingle et al., 1998; Pihan et al., 2003).

The failure of chromosomes to attach to the spindle also causes aneuploidy. These failures arise when kinetochore microtubules cannot form stable connections to chromosomes, or when they bind to more than one chromosome (Cimini and Degrassi, 2005). Some attachment defects are sensed by the spindle assembly checkpoint, but a tumor suppressor mutation may inactivate normal checkpoint signaling and allow mitosis to continue despite these mistakes. Chromosome misattachments are seen in many tumor types and are thought by many investigators to facilitate tumorigenesis.

Fig. 3.

Mitotic defects leading to whole-chromosome aneuploidy. (A) An anaphase cell that has undergone proper chromosome segregation and begun anaphase. (B–D) Tumor suppressor-associated mitotic spindle defects. (B) Individual chromosomes can segregate incorrectly owing to failed attachments of kinetochore microtubules. (C) Multiple chromosomes can segregate to the wrong daughter cell owing to tripolar spindle formation. (D) Cytokinesis can fail, resulting in a single, tetraploid daughter cell.

Fig. 3.

Mitotic defects leading to whole-chromosome aneuploidy. (A) An anaphase cell that has undergone proper chromosome segregation and begun anaphase. (B–D) Tumor suppressor-associated mitotic spindle defects. (B) Individual chromosomes can segregate incorrectly owing to failed attachments of kinetochore microtubules. (C) Multiple chromosomes can segregate to the wrong daughter cell owing to tripolar spindle formation. (D) Cytokinesis can fail, resulting in a single, tetraploid daughter cell.

Close modal

A final source of aneuploidy is the failure of cytokinesis following proper or improper chromosome segregation. Cytokinesis requires microtubules for both specifying the cytokinesis plane and delivering membrane components to the correct cortical site (Barr and Gruneberg, 2007). Failure to complete cytokinesis produces a single, tetraploid daughter cell. Tetraploidy, in turn, accelerates other genetic changes (Ganem et al., 2007).

The possible cellular outcomes following chromosome segregation errors include cell death, survival as an aneuploid cell and evolution with further genomic instability. Because many tumor suppressors also promote apoptosis signaling, their inactivation can lead to the dangerous combination of aneuploidy without appropriate apoptosis. The tumor suppressors discussed in this review have all been associated with one or more of these mitotic errors. Thus, their inactivation contributes to the overarching role of genomic instability in promoting cancer development.

In addition, since mitotic fidelity is so sensitive to microtubule destabilization, it may be impaired by heterozygosity for a microtubule-interacting tumor suppressor, a state that is not usually thought to promote cancer development. Such a role has been proposed for APC and may apply to other tumor suppressors as well (Nowak et al., 2002).

Spindle misorientation may increase the pool of tumor stem cells

A final mitotic defect caused by impaired astral microtubule function is spindle misorientation within the cell. The spindle orientation axis sets the cell division plane, which determines daughter cell inheritance and positioning. Asymmetric partitioning is usually seen in stem cell compartments and is considered to be a means of replenishing the stem cell pool. During development and injury/regeneration, a shift to symmetric division allows the stem cell compartment to expand (Morrison and Kimble, 2006). A similar switch in a tumor might also expand the stem cell population, impacting chemotherapy resistance and metastatic potential. Loss of astral microtubule attachments to the cell cortex could revert asymmetric spindle orientation to a symmetric pattern or could randomize spindle orientation, thereby increasing the cancer stem cell pool (Morrison and Kimble, 2006).

In summary, microtubule destabilization has the potential to alter some fundamental cellular processes that are known to be abnormal in cancer. These include altering cell growth and death signaling pathways, reducing cell polarity, increasing EMT-like migration, causing chromosome segregation errors, and producing spindle misorientation with expansion of the stem cell pool. Mutation of a microtubule interacting tumor suppressor would not need to cause all of these defects simultaneously; even one or two of these effects could send cell polarity or mitosis off track.

Despite their diverse structures and other cellular activities, several tumor suppressors with known associations to human cancer also stabilize microtubules. Reduced microtubule stability and its consequences, caused by inactivation of these proteins, may contribute to the evolution of a large percentage of human cancers.

APC

APC was initially identified as the disease gene for familial adenomatous polyposis (FAP), an inherited syndrome of early-onset colorectal polyposis (Nishisho et al., 1991). These patients develop hundreds of colonic polyps, some of which inevitably progress to carcinoma (Fodde and Khan, 1995). Families with germline APC mutations also exhibit extra-intestinal manifestations, including malignant gastrointestinal and nervous system tumors, benign congenital hypertrophy of the retinal pigment epithelium (CHRPE), osteomas, and dental and skin abnormalities. Somatic mutation of APC was subsequently shown to account for over 80% of sporadic colorectal cancers (Bodmer et al., 1989). Mutation of APC thus accounts for tens of thousands of colorectal tumors annually.

The APC gene encodes a ∼300-kDa protein that is responsible for cell growth, cell survival, DNA repair and RNA trafficking, as described in many comprehensive reviews (Aoki and Taketo, 2007; Fearnhead et al., 2001; Nathke, 2000; Nathke, 2004; Rusan and Peifer, 2008; Sieber et al., 2001). There is a second APC homolog, APC2, which has been suggested also to have a role in tumor suppression (Jarrett et al., 2001; van Es et al., 1999). Most of the APC mutations identified from FAP families and sporadic cases are clustered in central repeat regions of APC that bind to β-catenin. APC downregulates β-catenin and prevents uncontrolled growth by reducing the transcription of c-Myc and cyclin D (He et al., 1998; Rubinfeld et al., 1996; Tetsu and McCormick, 1999; Wong and Pignatelli, 2002).

APC-truncating mutations also remove carboxy-terminal APC regions that bind to cytoskeletal components (Hanson and Miller, 2005; Nathke, 2004; Nathke, 2005). APC-microtubule interactions include the direct binding of APC to microtubules by a basic region of APC, binding of APC to the microtubule plus-end tracking protein EB1, binding of APC to the microtubule-destabilizing protein mitotic centromere-associated kinase (MCAK), and binding of APC to microtubule-based kinesin motors (Banks and Heald, 2004; Deka et al., 1998; Jimbo et al., 2002; Munemitsu et al., 1994; Smith et al., 1994; Su et al., 1995). Phosphorylation of APC by glycogen synthase kinase 3β (GSK3β) reduces its affinity for microtubules, just as phosphorylation of traditional MAPs reduces their levels of microtubule binding (Zumbrunn et al., 2001). APC is a potent microtubule stabilizer, and the addition of the C-terminal regions that are lost upon tumor-associated mutation was found to dramatically increase microtubule polymerization and stability in an in vitro assay (Deka et al., 1998; Munemitsu et al., 1994; Nakamura et al., 2001; Zumbrunn et al., 2001). APC also localizes to centrosomes, although its function there is less well studied (Louie et al., 2004).

Because most tumorigenic APC mutations simultaneously abrogate APC–β-catenin interactions and APC-microtubule binding, it has been difficult to separate these roles experimentally. A mouse harboring a truncating APC mutation distal to the β-catenin binding regions (Apc1638T) showed developmental abnormalities but did not spontaneously form tumors (Smits et al., 1999). By contrast, human patients with distal APC mutations that preserve β-catenin interactions but eliminate binding to cytoskeletal components develop attenuated polyposis (Resta et al., 2001). The existence of these kindreds suggests that the loss of APC-cytoskeletal interactions may be sufficient for tumorigenesis.

Intestinal epithelial cells use a form of sheet migration, maintaining apico-basal polarization as they move upward from the crypt base. Microtubules deliver APC to the leading edges of migrating cells (Mimori-Kiyosue et al., 2000; Nathke et al., 1996). In a fibroblast scratch wound assay, APC stabilized a population of microtubule ends facing the leading edges of migrating cells; in addition, cells that were transfected with tumor-associated APC mutants showed increased migration through Transwell filters (Wen et al., 2004). Forced expression of full-length APC in mouse intestines caused changes in crypt morphology that were attributed to excessive cell migration (Munemitsu et al., 1994; Wong et al., 1996). Cell migration was reduced in intestinal crypts in which APC was inactivated and in cultured cells in which APC was eliminated (Kroboth et al., 2007; Munemitsu et al., 1994; Sansom et al., 2004). These results are all consistent with a role for APC in enhancing cell migration. Cell migration depends heavily on actin polymerization at the leading edge and APC interacts with several actin-associated proteins (Iizuka-Kogo et al., 2005; Mimori-Kiyosue et al., 2007; Watanabe et al., 2004; Wen et al., 2004). Thus, APC may mediate an important microtubule-actin association during the migration process.

APC also plays a major role in chromosome segregation. Studies correlating mutations in APC with aneuploidy and CIN in tumors and cancer cell lines have shown various degrees of association (Kearney et al., 1993; Tighe et al., 2001). A role for APC mutations in impairing chromosome segregation was shown in embryonic stem cells expressing truncated APC as the sole form of the protein in the cell (Fodde et al., 2001; Kaplan et al., 2001). Based on cell culture studies, various models for the mechanics of chromosome segregation defects have been proposed, including interference with the spindle assembly checkpoint, impaired chromosome attachments to kinetochore microtubules, and a failure to correct chromosome defects (Caldwell et al., 2007; Dikovskaya et al., 2007; Draviam et al., 2006; Green et al., 2005; Zhang et al., 2007; Zhang et al., 2009). These ideas are thoughtfully discussed in a recent review (Rusan and Peifer, 2008).

APC also plays a role in orienting the spindle within the cell (Caldwell et al., 2007; Fleming et al., 2009). In fly stem cells, APC was needed for asymmetric spindle orientation and was proposed to limit the size of the stem cell compartment. In cultured mammalian tumor cells, APC RNA interference (RNAi) induced astral microtubule depolymerization, causing spindle misorientation and cytokinesis failure (Caldwell et al., 2007). In mouse intestines, the presence of mutant APC led to spindle misorientation without reducing astral microtubule length, suggesting a role in astral microtubule attachment to the cell cortex (Fleming et al., 2009). In summary, APC stabilizes microtubules and links the microtubule and actin polymer systems. Its inactivation disrupts cell migration, chromosome segregation and mitotic spindle orientation.

RASSF1A

Although no familial syndromes involving mutation of the RASSF1 gene have been identified, epigenetic inactivation of the A isoform of RASSF1 is one of the most common molecular events in human cancer (Donninger et al., 2007). Both promoter hypermethylation and somatic mutations have been described; hypermethylation of the RASSF1A promoter is well documented in small cell and non-small cell lung cancer and other solid tumors (Donninger et al., 2007). Strong evidence for the causative role of this epigenetic inactivation in cancer was provided by the finding that targeted deletion of Rassf1a in mice increases the incidence of many types of spontaneous, radiation- and carcinogen-induced tumors (Tommasi et al., 2005; van der Weyden et al., 2005).

RASSF1A is one of seven isoforms of the RASSF1 gene (RASSF1A-G), which itself belongs to a family of 10 genes (RASSF1-RASSF10). The RASSF1A isoform encodes a 39-kDa protein with a C-terminal Ras association domain (Donninger et al., 2007). It acts as a Ras effector, which may require heterodimerization with the related NORE1 (later designated as RASSF5) protein (Ortiz-Vega et al., 2002; Richter et al., 2009). Another major function associated with RASSF1A is regulation of the G1-S cell cycle transition through interactions with the c-Jun N-terminal kinase (JNK) pathway, the transcription factor p120E4F or cyclin D1 (Donninger et al., 2007). RASSF1A also acts as a pro-apoptotic factor, interacting with several apoptotic pathway components (Richter et al., 2009).

RASSF1A stabilizes microtubules. Green fluorescent protein (GFP)-RASSF1A colocalized and co-sedimented with microtubules from cell extracts, and some cancer-associated mutations of RASSF1A abolished its microtubule localization (Dallol et al., 2004; Liu et al., 2003; Rong et al., 2004). A microtubule-binding region of the RASSF1A protein has been mapped, and interaction with the microtubule-stabilizing protein MAP1B has been reported (Dallol et al., 2004; Liu et al., 2003). Overexpression of GFP-RASSF1A increased microtubule stability and prevented microtubule depolymerization by drug or cold treatment, and RASSF1A null fibroblasts were more sensitive than controls to anti-microtubule drugs (Liu et al., 2003; Rong et al., 2004; Vos et al., 2004).

Interestingly, the addition of RASSF1A to RASSF1A null cells reduced migration in a wound-healing assay, whereas RASSF1A RNAi increased cellular migration (Dallol et al., 2005). Both of these results are consistent with a role for the protein in preventing cell migration, which is the opposite effect from that seen for APC.

In mitotic cells, enhanced GFP (EGFP)-RASSF1A localized to spindle microtubules and it was retained at the spindle poles in the presence of the microtubule-depolymerizing agent nocodazole (Liu et al., 2003). Antibody staining showed the spindle pole but not the spindle microtubule association, suggesting that lower affinity microtubule binding may require overexpression of RASSF1A (Guo et al., 2007; Liu et al., 2008). RASSF1A overexpression caused an increase in monopolar spindles and cytokinesis failure (Guo et al., 2007; Liu et al., 2003), whereas RASSF1A RNAi caused a premature exit from mitosis, with multipolar spindles and misaligned and lagging chromosomes (Liu et al., 2003; Song et al., 2004). As a possible mechanism, RASSF1A is both an activator of, and a substrate for, the mitotic kinase Aurora A, which controls centrosome separation and is itself frequently upregulated in tumors (Rong et al., 2007). In support of this, a RASSF1A mutation that mimicked Aurora phosphorylation bypassed the mitotic delay caused by Aurora A RNAi (Song et al., 2009).

In summary, RASSF1A, like APC, stabilizes microtubules. Unlike APC, RASSF1A appears to be an inhibitor of cell migration. Its loss causes pleiotropic effects on the mitotic spindle, which could be mediated by either microtubule destabilization or effects on Aurora kinases, or both.

VHL

The VHL gene is mutated in the germline of patients with familial von Hippel-Lindau syndrome, which is characterized by renal cell cancer, malignancies of the cerebellum and retina, pheochromocytomas, visceral cysts and other tumors (Kaelin, 2008). Loss of heterozygosity at the VHL locus and VHL promoter methylation were subsequently shown to be responsible for a large number of sporadic kidney cancers (Nyhan et al., 2008). Thus, VHL fulfills a gatekeeper role in the renal epithelium similar to that of APC in the intestine.

VHL encodes a protein of 30 kDa (VHL30), with translation from an internal initiation codon producing a 19-kDa isoform (VHL19) with a truncated amino terminus. Both isoforms are often discussed together in publications, making it difficult to interpret the individual functions of the two isoforms. VHL acts as the substrate recognition component of an E3 ubiquitin ligase that promotes degradation of the transcription factor hypoxia-inducible factor 1α (HIF1α) in the presence of oxygen (Kaelin, 2007; Nyhan et al., 2008). Cells lacking VHL behave as if they were experiencing hypoxia: they upregulate HIF1, leading to expression of pro-angiogenic factors such as vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF).

As with APC and RASSF1A, VHL stabilizes microtubules. A decade after its discovery, an immunofluorescence study using a novel antibody revealed that VHL is localized to microtubules (Hergovich et al., 2003). It is not clear why earlier studies failed to show this localization using other antibodies and GFP fusions. Potential explanations include the frequent use of immunohistochemistry (IHC, which is inadequate for imaging microtubules) and the possibility that C-terminal GFP fusions that did not localize correctly. Equally important may be a distinction between the two VHL isoforms, as only the less-abundant VHL30 appears to localize to microtubules (Hergovich et al., 2003). VHL30 was observed as being bound to microtubules in a co-pelleting assay; this binding was later shown to be mediated by the kinesin Kif3A (Hergovich et al., 2003; Lolkema et al., 2007). VHL stabilizes microtubules that are exposed to the microtubule-depolymerizing drug nocodazole, an ability that is abrogated by disease-associated VHL mutations (Hergovich et al., 2003; Hergovich et al., 2006). Fluorescence recovery after photobleaching (FRAP) experiments suggested that VHL might reduce the turnover of microtubules at the periphery of cultured cells (Lolkema et al., 2004). This ability of VHL to stabilize microtubules depends on two serine residues in VHL30 that are substrates for phosphorylation by GSK3β (Hergovich et al., 2006). In this way, VHL is similar to APC in interacting with GSK3β and similar to other microtubule-stabilizing proteins whose activities are reduced by GSK3β phosphorylation.

VHL was recently found to be required for the microtubule-based process of ciliogenesis, which is especially important in renal development, and to bind to the ciliary kinesin-2 complex (Kuehn et al., 2007; Lolkema et al., 2007; Schermer et al., 2006). A failure to support ciliogenesis correlates with disease mutations (Lutz and Burk, 2006; Thoma et al., 2007), and loss of VHL causes ciliary depletion, but only when GSK3β is also inhibited (Thoma et al., 2007). Loss of ciliogenesis might play a role in tumor development by disrupting cell polarity and altering tissue architecture.

An elevation of HIF1 caused by VHL inactivation also represses the expression of the crucial cell-cell junction protein E-cadherin, which is itself a microtubule-interacting tumor suppressor. Increased levels of HIF also repress the expression of the tight junction components occludin and claudin (Calzada et al., 2006; Evans et al., 2007; Harten et al., 2009). The resulting loss of adherens and tight junctions may reduce epithelial polarity and contribute to an EMT-like process, further reducing microtubule stability.

A role for VHL in mitotic spindle dynamics was discovered recently. The depletion of VHL by RNAi caused chromosome segregation defects leading to whole-chromosome aneuploidy; this correlated with a reduction of the mitotic checkpoint protein Mad2 (Evans et al., 2007). VHL RNAi also caused mitotic spindle misorientation, which correlated with the loss of microtubule-stabilizing regions of the protein, and was associated with shortened astral microtubules (Evans et al., 2007).

In summary, VHL acts as a microtubule-stabilizing protein, both in the cytoplasm and the primary cilia. It maintains E-cadherin and tight junction protein levels and hence cell-cell attachment, contributing to epithelial polarity. It protects cells from aneuploidy and orients the mitotic spindle, thus potentially regulating genomic stability and the size of the stem cell pool.

E-cadherin

Hereditary mutation in the CDH1 gene that encodes E-cadherin causes the rare hereditary diffuse gastric cancer (HDGC) syndrome (Dunbier and Guilford, 2001). These germline mutations account for about a third of hereditary gastric cancers and 1% of gastric cancers overall (Dunbier and Guilford, 2001). Affected patients are also predisposed to lobular breast cancer, and recently, a family with lobular breast cancer, but without gastric cancer, was found to harbor an E-cadherin mutation (Masciari et al., 2007). Highlighting its role in lobular breast cancer pathogenesis, E-cadherin mutations have been seen in the precursor lesion lobular carcinoma in situ (LCIS), and reduced E-cadherin expression is used clinically to help distinguish between ductal and lobular breast cancers (Cowin et al., 2005; Lerwill, 2004). Epigenetic inactivation of E-cadherin is common in multiple cancer types (Strathdee, 2002).

E-cadherin is the epithelially expressed member of the cadherin gene family, which includes many additional genes that encode cadherin proteins (Halbleib and Nelson, 2006). These proteins include N-cadherin, which is implicated in EMT-like processes. E-cadherin encodes a single-pass transmembrane protein of ∼120 kDa that mediates calcium-dependent homotypic adhesion between epithelial cells (van Roy and Berx, 2008). Cadherins are needed for the formation of both adherens junctions and tight junctions between cells. Their intracellular domains coordinate actin polymerization through α-catenin; these domains also stimulate signaling pathways, the best studied of which is the regulation of Wnt signaling through interactions with β-catenin.

An appreciation of the interactions of E-cadherin with microtubules is beginning to emerge (Hall, 2009; Stehbens et al., 2009). The formation of E-cadherin-dependent cell contacts is associated with a dramatic reorganization of microtubules and changes in their dynamic properties (Bre et al., 1990; Buendia et al., 1990; Wadsworth and Bottaro, 1996). Cadherins and their associated junctional complexes can substitute for centrosomes in their ability to stabilize microtubule minus ends (Chausovsky et al., 2000). Recently, E-cadherin-dependent adherens junctions were shown to anchor microtubule minus ends and, possibly, to nucleate new microtubules at these junctions, with implications for trafficking molecules towards the junctions (Meng et al., 2008). The stabilization of minus ends converts the dominant microtubule behavior from treadmilling (continuous minus-end depolymerization coupled to plus-end polymerization) into plus-end dynamic instability (Chausovsky et al., 2000). Adherens junctions also interact independently with microtubule plus ends to enhance their stability (Waterman-Storer et al., 2000).

Loss of E-cadherin-mediated cell adhesion is thought to be a crucial step in the EMT, presumed to be a prerequisite for the development of metastasis (Schmalhofer et al., 2009). Many metastasis-associated pathways converge on the downregulation of E-cadherin expression (Cano et al., 2000; Guarino et al., 2007). Loss of E-cadherin is often most noticeable at the tumor front, where cells egress from the tumor.

A role for E-cadherin in mediating spindle orientation was described recently. A dominant negative form of E-cadherin caused spindle misorientation without disrupting epithelial junctions or polarity, but E-cadherin RNAi misoriented spindles only when another cadherin (cadherin 6) was also eliminated (den Elzen et al., 2009). Altogether, E-cadherin plays bidirectional roles in enhancing the stability of microtubule plus and minus ends, maintaining epithelial polarity, and orientating the mitotic spindle.

LKB1

LKB1, which is also called serine/threonine kinase 11 (STK11), is mutated in the familial Peutz-Jeghers syndrome (PJS). Affected individuals develop mucocutaneous pigmentation and multiple, benign hamartomatous intestinal polyps. The incidence of colorectal cancer in PJS patients is independently amplified by many-fold, and cancers of other sites including the breast, ovary, lung and testes are also increased (Hearle et al., 2006). Somatic LKB1 mutations have been identified in a subset of sporadic non-small cell lung cancers, especially in male smokers, as well as in a variety of other sporadic tumors (Giardiello et al., 2000; Ji et al., 2007; Matsumoto et al., 2007). LKB1 is thus a key tumor suppressor in several tissues.

LKB1 encodes an ∼50-kDa protein with serine-threonine kinase activity (Hezel and Bardeesy, 2008). It phosphorylates downstream targets that are involved in two separate, yet linked, activities: energy utilization and cell polarity (Hezel and Bardeesy, 2008). LKB1 activates several members of the AMP-activated protein kinase (AMP kinase) family, shifting the cell from a state of ATP consumption to one of ATP production. The C. elegans homolog of LKB1 was identified as partitioning defective gene 4 (Par4) in a screen for mutants that failed to properly carry out asymmetric cell division in the developing embryo (Watts et al., 2000). Mammalian LKB1 is the only protein that has been shown to cause single cells to autonomously generate apico-basal polarity in the absence of cell-cell contacts (Baas et al., 2004). The AMP kinases that are downstream of LKB1 phosphorylate the mammalian target of rapamycin (mTOR), accelerating tight junction formation. Together, the overall activity of LKB1 is to increase energy production and to enhance epithelial cell polarization.

In addition to its effects on cell polarity, LKB1 was demonstrated to regulate microtubule stability through effects on proteins that control microtubule dynamics. LKB1 phosphorylates the microtubule affinity-regulating kinases (MARKs), which in turn phosphorylate MAPs such as the stabilizing protein Tau (Kojima et al., 2007). In vitro, a cascade of phosphorylation from LKB1 to MARK2 to Tau reduced initial rates of microtubule polymerization (Kojima et al., 2007). In cultured cells, the expression of LKB1 did not alter the microtubule array, but it did suppress microtubule re-growth following nocodazole washout (Kojima et al., 2007). These results suggest that LKB1 reduces microtubule stability, which is opposite to the activity of the other tumor suppressors discussed thus far.

LKB1 also influences cell division. It colocalizes with meiotic spindles in mouse oocytes, which are tethered to the oocyte cortex (Szczepanska and Maleszewski, 2005). Mutations of the fly LKB1 homolog cause structural defects in the mitotic spindle, including polyploidy, improperly segregated chromosomes, reduced astral microtubule density, and loss of the normal asymmetry of spindle orientation – all of these phenotypes are consistent with reduced stability of spindle microtubules (Bonaccorsi et al., 2007; Lee et al., 2007). In summary, LKB1 differs from the other tumor suppressors in having more complex effects on microtubules, but it has similar roles in promoting epithelial polarization and in protecting mitotic spindle dynamics.

Neurofibromin and merlin

The neurofibromatoses are familial cancer syndromes caused by the mutation of two tumor suppressors, neurofibromatosis 1 and 2 (NF1 and NF2), which encode neurofibromin and merlin (also called schwannomin), respectively. These syndromes are associated with benign tumors of the nervous system, pigmentary lesions, malignant tumors of the peripheral nerve sheath (MPNST), gliomas and myelodysplastic syndromes of the blood (Reed and Gutmann, 2001). NF2 mutations have been found in sporadic schwannomas, meningiomas, melanomas and mesotheliomas (Reed and Gutmann, 2001; Xiao et al., 2003).

The NF1 gene product, neurofibromin, is a 280-kDa protein that acts as a GTPase activating protein (GAP) for Ras. When neurofibromin function is reduced, Ras remains in an active state that promotes cell proliferation through growth factor pathways and MAP kinase signaling cascades (Trovo-Marqui and Tajara, 2006). The NF2 gene product, merlin, is a 65-kDa protein with homology to the ezrin, radixin and moesin (ERM) family of proteins, which organize plasma membranes and link them to underlying cortical actin (McClatchey and Fehon, 2009; Scoles, 2008). Merlin anchors the actin cytoskeleton to the overlying membrane (Xu and Gutmann, 1998). Similar to neurofibromin, it also controls cell proliferation through a Ras mechanism. Merlin also regulates the Rho GTPase Rac, which is involved in cell-cell and cell-matrix adhesion (Xiao et al., 2003). Finally, merlin was suggested to play a role in stabilizing adherens junctions, which are needed for epithelial cell polarization (Lallemand et al., 2003). Thus, these proteins contribute to epithelial polarity.

Merlin localizes to microtubules by immunofluorescence (Gregory et al., 1993) and binds to microtubules in an in vitro assay (Muranen et al., 2007). The phosphorylation of merlin is thought to unfold it and promote its microtubule binding (Muranen et al., 2007; Scoles, 2008; Xu and Gutmann, 1998). Both neurofibromin and merlin have been co-purified from HeLa cells in large complexes with microtubule-associated kinesin-1 (Hakimi et al., 2002). Recombinant merlin increased tubulin polymerization in an in vitro assay, and merlin increased microtubule turnover as assessed by FRAP (Muranen et al., 2007). Roles for neurofibromin and merlin in the microtubule-based transport of vesicular cargoes have been proposed and remain to be tested (Hakimi et al., 2002).

Centrosome-interacting tumor suppressors: p53 and BRCA1

Two important tumor suppressor proteins, p53 and breast cancer 1 (BRCA1), bind to centrosomes and repress centrosome duplication. Their loss can promote centrosome overduplication, which can in turn lead to spindle multipolarity and aneuploidy.

Hereditary mutation of p53 causes the Li-Fraumeni cancer syndrome, and p53 mutations are among the most common cancer-associated mutations (Malkin, 1994). In addition to its well-known roles in controlling cell death and cell cycle progression, the p53 gene product also controls centrosome number. Cells lacking p53 undergo abnormal centrosome amplification by two mechanisms: cell cycle disruption and physical interaction with centrosomes (Fukasawa et al., 1996; Shinmura et al., 2007). The physical interaction prevents centrosome overduplication independently of p53 binding to DNA (Tarapore et al., 2001). Interestingly, in prolonged culture, p53 mutant cells eventually suppress this centrosome amplification, suggesting that the selective benefit of the abnormality might be transient (Chiba et al., 2000). Finally, p53 has been noted to localize to mitotic spindle poles, but defects in mitotic spindle dynamics owing to a loss of p53 have not been demonstrated (Morris et al., 2000; Tritarelli et al., 2004).

Germline mutation of the breast and ovarian cancer tumor suppressor gene BRCA1 accounts for the familial breast and ovarian cancer syndrome. In addition to its major role in DNA repair, BRCA1 also regulates centrosome duplication. BRCA1 binds to centrosomes and mitotic spindle poles (Hsu and White, 1998); its structural partners at the centrosome include the microtubule-nucleating protein γ-tubulin and proteins that control mitotic spindle assembly (Hsu and White, 1998; Joukov et al., 2006; Sankaran et al., 2007). BRCA1 ubiquitinates γ-tubulin and prevents its over-recruitment to the centrosome (Sankaran et al., 2007; Starita et al., 2004). In its absence, cells develop supernumerary centrosomes that show an increased microtubule-nucleating capacity (Starita et al., 2004; Xu et al., 1999). This effect may be tissue specific, as it could not be produced in non-breast cell lines (Starita et al., 2004). The depletion of BRCA1 also reduced the mitotic spindle pole focusing that was induced by activated Ran in Xenopus egg extracts, a centrosome-independent form of spindle assembly (Joukov et al., 2006). These effects on spindles have the potential to promote aneuploidy.

Ultimately, it will be useful to know whether we can exploit tumor suppressor interactions with microtubules to guide diagnostic and therapeutic practice. If changes in microtubule stability impact the evolution of a pre-neoplastic lesion to an invasive cancer, or alter the metastatic potential of a tumor, this information could inform decisions about how aggressively to treat a patient. Easy diagnostic readouts of microtubule stability will be needed, as current biochemical assays of microtubule polymerization require fresh material, and microtubule imaging depends on sophisticated optics and special sample handling. Assaying surrogates for microtubule destabilization may be easier and more practical than assaying microtubules directly.

We also do not know whether defective microtubule regulation predicts the response of a tumor to chemotherapy. Anti-microtubule drugs are one of the largest and most effective classes of chemotherapeutic drugs. Many of the currently used anti-microtubule drugs, such as paclitaxel and vinca alkaloids, are subject to multidrug resistance efflux, further complicating the interpretation of their effects relative to microtubule regulation (Morris and Fornier, 2008). This may soon change with the introduction of the epothilones and other compounds that are not subject to drug efflux (Morris and Fornier, 2008). The ability to predict the effects of these drugs on tumor cells based on measures of existing microtubule stability in the tumor would be a major advance.

Anti-microtubule drugs are thought to work by causing mitotic arrest followed by cell death. If all anti-microtubule drugs acted by this same pathway, however, one would expect similar effects on cell death, and these are not always seen. Thus, the connections between microtubule stability and the cell death machinery need to be better understood.

Alterations in microtubule stability could either synergize with, or antagonize, anti-microtubule drugs, and mitotic spindle defects could be an asset or a liability for cell survival (Chandhok and Pellman, 2009). Recent work has begun to demonstrate that, following mitotic arrest, cell fate decisions are not only cell type and drug type specific, but also vary within a clonal cell population that has been exposed to the same drug (Brito and Rieder, 2008; Gascoigne and Taylor, 2008; Orth et al., 2008). It will thus be interesting to determine whether the response of cancer cells to anti-microtubule drugs depends on existing microtubule defects induced by tumor suppressor mutations.

Just as tumor suppressor mutations may influence the response of a cell to anti-microtubule drugs, treatment with these drugs may alter the function of a microtubule-bound tumor suppressor. Experiments to determine interactions between anti-microtubule drugs and microtubule-interacting tumor suppressors could help to narrow the gap between a fascinating biological problem and more individualized cancer therapy.

We thank Vladimir Rodionov, Mimi Shirasu-Hiza and anonymous reviewers for valuable comments on the manuscript.

Acilan
C
,
Saunders
WS
(
2008
).
A tale of too many centrosomes
.
Cell
134
,
572
575
.
Amin
N
,
Khan
A
,
St Johnston
D
,
Tomlinson
I
,
Martin
S
,
Brenman
J
,
McNeill
H
(
2009
).
LKB1 regulates polarity remodeling and adherens junction formation in the Drosophila eye
.
Proc Natl Acad Sci USA
106
,
8941
8946
.
Aoki
K
,
Taketo
MM
(
2007
).
Adenomatous polyposis coli (APC): a multifunctional tumor suppressor gene
.
J Cell Sci
.
120
,
3327
3335
.
Baas
AF
,
Kuipers
J
,
van der Wel
NN
,
Batlle
E
,
Koerten
HK
,
Peters
PJ
,
Clevers
HC
(
2004
).
Complete polarization of single intestinal epithelial cells upon activation of LKB1 by STRAD
.
Cell
116
,
457
466
.
Banks
JD
,
Heald
R
(
2004
).
Adenomatous polyposis coli associates with the microtubule-destabilizing protein XMCAK
.
Curr Biol
.
14
,
2033
2038
.
Barr
FA
,
Gruneberg
U
(
2007
).
Cytokinesis: placing and making the final cut
.
Cell
131
,
847
860
.
Bilder
D
,
Li
M
,
Perrimon
N
(
2000
).
Cooperative regulation of cell polarity and growth by Drosophila tumor suppressors
.
Science
289
,
113
116
.
Bodmer
WF
,
Cottrell
S
,
Frischauf
AM
,
Kerr
IB
,
Murday
VA
,
Rowan
AJ
,
Smith
MF
,
Solomon
E
,
Thomas
H
,
Varesco
L
(
1989
).
Genetic analysis of colorectal cancer
.
Int Symp Princess Takamatsu Cancer Res Fund
20
,
49
59
.
Bonaccorsi
S
,
Mottier
V
,
Giansanti
MG
,
Bolkan
BJ
,
Williams
B
,
Goldberg
ML
,
Gatti
M
(
2007
).
The Drosophila Lkb1 kinase is required for spindle formation and asymmetric neuroblast division
.
Development
134
,
2183
2193
.
Bre
MH
,
Pepperkok
R
,
Hill
AM
,
Levilliers
N
,
Ansorge
W
,
Stelzer
EHK
,
Karsenti
E
(
1990
).
Regulation of microtubule dynamics and nucleation during polarization in MDCK II Cells
.
J Cell Biol
.
111
,
3013
3021
.
Brito
DA
,
Rieder
CL
(
2008
).
The ability to survive mitosis in the presence of microtubule poisons differs significantly between human nontransformed (RPE-1) and cancer (U2OS, HeLa) cells
.
Cell Motil Cytoskeleton
66
,
437
447
.
Buendia
B
,
Bre
M
,
Griffiths
G
,
Karsenti
E
(
1990
).
Cytoskeletal control of centrioles movement during the establishment of polarity in madin-darby canine kidney cells
.
J Cell Biol
.
110
,
1123
1135
.
Caldwell
CM
,
Green
RA
,
Kaplan
KB
(
2007
).
APC mutations lead to cytokinetic failures in vitro and tetraploid genotypes in Min mice
.
J Cell Biol
.
178
,
1109
1120
.
Calzada
MJ
,
Esteban
MA
,
Feijoo-Cuaresma
M
,
Castellanos
MC
,
Naranjo-Suárez
S
,
Temes
E
,
Méndez
F
,
Yánez-Mo
M
,
Ohh
M
,
Landázuri
MO
(
2006
).
von Hippel-Lindau tumor suppressor protein regulates the assembly of intercellular junctions in renal cancer cells through hypoxia-inducible factor independent mechanisms
.
Cancer Res
.
66
,
1553
1560
.
Cano
A
,
Pérez-Moreno
MA
,
Rodrigo
I
,
Locascio
A
,
Blanco
MJ
,
del Barrio
MG
,
Portillo
F
,
Nieto
MA
(
2000
).
The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression
.
Nat Cell Biol
.
2
,
76
83
.
Chandhok
NS
,
Pellman
D
(
2009
).
A little CIN may cost a lot: revisiting aneuploidy and cancer
.
Curr Opin Genet Dev
.
19
,
74
81
.
Chausovsky
A
,
Bershadsky
AD
,
Borisy
GG
(
2000
).
Cadherin-mediated regulation of microtubule dynamics
.
Nat Cell Biol
.
2
,
797
804
.
Chiba
S
,
Okuda
M
,
Mussman
JG
,
Fukasawa
K
(
2000
).
Genomic convergence and suppression of centrosome hyperamplification in primary p53−/− cells in prolonged culture
.
Exp Cell Res
.
258
,
310
321
.
Cimini
D
,
Degrassi
F
(
2005
).
Aneuploidy: a matter of bad connections
.
Trends Cell Biol
.
15
,
442
451
.
Cowin
P
,
Rowlands
TM
,
Hatsell
SJ
(
2005
).
Cadherins and catenins in breast cancer
.
Curr Opin Cell Biol
.
17
,
499
508
.
Dallol
A
,
Agathanggelou
A
,
Fenton
SL
,
Ahmed-Choudhury
J
,
Hesson
L
,
Vos
MD
,
Clark
GJ
,
Downward
J
,
Maher
ER
,
Latif
F
(
2004
).
RASSF1A interacts with microtubule-associated proteins and modulates microtubule dynamics
.
Cancer Res
.
64
,
4112
4116
.
Dallol
A
,
Agathanggelou
A
,
Tommasi
S
,
Pfeifer
GP
,
Maher
ER
,
Latif
F
(
2005
).
Involvement of the RASSF1A tumor suppressor gene in controlling cell migration
.
Cancer Res
.
65
,
7653
7659
.
Deka
JR
,
Kuhlmann
JR
,
Müller
O
(
1998
).
A domain within the tumor suppressor protein APC shows very similar biochemical properties as the microtubule-associated protein tau
.
Eur J Biochem
.
253
,
591
597
.
den Elzen
N
,
Buttery
CV
,
Maddugoda
MP
,
Ren
G
,
Yap
AS
(
2009
).
Cadherin adhesion receptors orient the mitotic spindle during symmetric cell division in mammalian epithelia
.
Mol Biol Cell
20
,
3740
3750
.
Desai
A
,
Mitchison
TJ
(
1997
).
Microtubule polymerization dynamics
.
Annu Rev Cell Dev Biol
.
13
,
83
117
.
Dikovskaya
D
,
Schiffmann
D
,
Newton
IP
,
Oakley
A
,
Kroboth
K
,
Sansom
O
,
Jamieson
TJ
,
Meniel
V
,
Clarke
A
,
Näthke
IS
(
2007
).
Loss of APC induces polyploidy as a result of a combination of defects in mitosis and apoptosis
.
J Cell Biol
.
176
,
183
195
.
Donninger
H
,
Vos
MD
,
Clark
GJ
(
2007
).
The RASSF1A tumor suppressor
.
J Cell Sci
.
120
,
3163
3172
.
Draviam
VM
,
Shapiro
I
,
Aldridge
B
,
Sorger
PK
(
2006
).
Misorientation and reduced stretching of aligned sister kinetochores promote chromosome missegregation in EB1- or APC-depleted cells
.
EMBO J
.
25
,
2814
2827
.
Drewes
G
,
Ebneth
A
,
Mandelkow
EM
(
1998
).
MAPs, MARKs and microtubule dynamics
.
Trends Biochem Sci
.
23
,
307
311
.
Dugina
VB
,
Alexandrova
AY
,
Lane
K
,
Bulanova
E
,
Vasilie
JM
(
1995
).
The role of the microtubular system in the cell response to HGF/SF
.
J Cell Sci
.
108
,
1659
1667
.
Dunbier
A
,
Guilford
P
(
2001
).
Hereditary diffuse gastric cancer
.
Adv Cancer Res
.
83
,
55
65
.
Etienne-Manneville
S
(
2008
).
Polarity proteins in migration and invasion
.
Oncogene
27
,
6970
6980
.
Evans
AJ
,
Russell
RC
,
Roche
O
,
Burry
TN
,
Fish
JE
,
Chow
VW
,
Kim
WY
,
Saravanan
A
,
Maynard
MA
,
Gervais
ML
, et al.
(
2007
).
VHL promotes E2 box-dependent E-cadherin transcription by HIF-mediated regulation of SIP1 and snail
.
Mol Cell Biol
.
27
,
157
169
.
Fearnhead
NS
,
Britton
MP
,
Bodmer
WF
(
2001
).
The ABC of APC
.
Hum Mol Genet
.
10
,
721
733
.
Flaiz
C
,
Utermark
T
,
Parkinson
DB
,
Poetsch
A
,
Hanemann
CO
(
2008
).
Impaired intercellular adhesion and immature adherens junctions in merlin-deficient human primary schwannoma cells
.
Glia
56
,
506
515
.
Fleming
ES
,
Temchin
M
,
Wu
Q
,
Maggio-Price
L
,
Tirnauer
JS
(
2009
).
Spindle misorientation in tumors from APCmin/+ mice
.
Mol Carcinog
.
48
,
592
598
.
Fodde
R
,
Khan
PM
(
1995
).
Genotype-phenotype correlations at the adenomatous polyposis coli (APC) gene
.
Crit Rev Oncog
.
6
,
291
303
.
Fodde
R
,
Kuipers
J
,
Rosenberg
C
,
Smits
R
,
Kielman
M
,
Gaspar
C
,
van Es
JH
,
Breukel
C
,
Wiegant
J
,
Giles
RH
, et al.
(
2001
).
Mutations in the APC tumour suppressor gene cause chromosomal instability
.
Nat Cell Biol
.
3
,
433
438
.
Fukasawa
K
,
Choi
T
,
Kuriyama
R
,
Rulong
S
,
Vande Woude
GF
(
1996
).
Abnormal centrosome amplification in the absence of p53
.
Science
271
,
1744
1747
.
Ganem
NJ
,
Storchova
Z
,
Pellman
D
(
2007
).
Tetraploidy, aneuploidy and cancer
.
Curr Opin Genet Dev
.
17
,
157
162
.
Gascoigne
KE
,
Taylor
SS
(
2008
).
Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs
.
Cancer Cell
14
,
111
122
.
Giardiello
FM
,
Brensinger
JD
,
Tersmette
AC
,
Goodman
SN
,
Petersen
GM
,
Booker
SV
,
Cruz-Correa
M
,
Offerhaus
JA
(
2000
).
Very high risk of cancer in familial peutz-jeghers syndrome
.
Gastroenterology
119
,
1447
1453
.
Green
RA
,
Wollman
R
,
Kaplan
KB
(
2005
).
APC and EB1 function together in mitosis to regulate spindle dynamics and chromosome alignment
.
Mol Biol Cell
16
,
4609
4622
.
Gregory
PE
,
Gutmann
DH
,
Mitchell
A
,
Park
S
,
Bugoski
M
,
Jacks
T
,
Wood
DL
,
Jove
R
,
Collins
FS
(
1993
).
Neurofibrosis type 1 gene product (neurofibromin) associates with microtubules
.
Somatic Cell Mol Genet
.
19
,
265
274
.
Guarino
M
,
Rubino
B
,
Ballabio
G
(
2007
).
The role of epithelial-mesenchymal transition in cancer pathology
.
Pathology
39
,
305
318
.
Guo
C
,
Tommasi
SL
,
Liu
L
,
Yee
JK
,
Dammann
R
,
Pfeifer
GP
(
2007
).
RASSF1A is part of a complex similar to the Drosophila Hippo/Salvador/Lats tumor-suppressor network
.
Curr Biol
.
17
,
700
705
.
Hahn
WC
,
Weinberg
RA
(
2002
).
Rules for making human tumor cells
.
N Engl J Med
.
347
,
1593
1603
.
Hakimi
M-A
,
Speicher
DW
,
Shiekhattar
R
(
2002
).
The motor protein Kinesin-1 Links neurofibromin and merlin in a common cellular pathway of neurofibromatosis
.
J Biol Chem
.
277
,
36909
36912
.
Halbleib
JM
,
Nelson
WJ
(
2006
).
Cadherins in development: cell adhesion, sorting, and tissue morphogenesis
.
Genes Dev
.
20
,
3199
3214
.
Hall
A
(
2009
).
The cytoskeleton and cancer
.
Cancer Metastasis Rev
.
28
,
5
14
.
Hammond
JW
,
Cai
D
,
Verhey
KJ
(
2008
).
Tubulin modifications and their cellular functions
.
Curr Opin Cell Biol
.
20
,
71
76
.
Hanahan
D
,
Weinberg
RA
(
2000
).
The hallmarks of cancer
.
Cell
100
,
57
70
.
Hanson
CA
,
Miller
JR
(
2005
).
Non-traditional roles for the adenomatous polyposis coli (APC) tumor suppressor protein
.
361
,
1
12
.
Harten
SK
,
Shukla
D
,
Barod
R
,
Hergovich
A
,
Balda
MS
,
Matter
K
,
Esteban
MA
,
Maxwell
PH
(
2009
).
Regulation of renal epithelial tight junctions by the von Hippel-Lindau tumor suppressor gene involves occludin and claudin 1 and is independent of E cadherin
.
Mol Biol Cell
20
,
1089
1101
.
He
TC
,
Sparks
AB
,
Rago
C
,
Hermeking
H
,
Zawel
L
,
da Costa
LT
,
Morin
PJ
,
Vogelstein
B
,
Kinzler
KW
(
1998
).
Identification of c-MYC as a Target of the APC Pathway
.
Science
281
,
1509
1512
.
Hearle
N
,
Schumacher
V
,
Menko
FH
,
Olshwang
S
,
Boardman
LA
,
Gille
JJ
,
Keller
JJ
,
Westerman
AM
,
Scott
RJ
,
Lim
W
, et al.
(
2006
).
Frequency and spectrum of cancers in the Peutz-Jeghers syndrome
.
Clin Cancer Res
.
12
,
3209
3215
.
Hergovich
A
,
Lisztwan
J
,
Barry
R
,
Ballschmieter
P
,
Krek
W
(
2003
).
Regulation of microtubule stability by the von Hippel-Lindau tumour suppressor protein pVHL
.
Nat Cell Biol
.
5
,
64
70
.
Hergovich
A
,
Lisztwan
J
,
Thoma
CR
,
Wirbelauer
C
,
Barry
RE
,
Krek
W
(
2006
).
Priming-dependent phosphorylation and regulation of the tumor suppressor pVHL by glycogen synthase kinase 3
.
Mol Biol Cell
26
,
5784
5796
.
Hezel
AF
,
Bardeesy
N
(
2008
).
LKB1; linking cell structure and tumor suppression
.
Oncogene
27
,
6908
6919
.
Howard
J
,
Hyman
AA
(
2003
).
Dynamics and mechanics of the microtubule plus end
.
Nature
422
,
753
758
.
Hsu
L-C
,
White
RL
(
1998
).
BRCA1 is associated with the centrosome during mitosis
.
Proc Natl Acad Sci USA
95
,
12983
12988
.
Iizuka-Kogo
A
,
Shimomura
A
,
Senda
T
(
2005
).
Colocalization of APC and DLG at the tips of cellular protrusions in cultured epithelial cells and its dependency on cytoskeletons
.
Histochem Cell Biol
.
123
,
67
73
.
Ivanov
AI
,
McCall
IC
,
Babbin
B
,
Samarin
SN
,
Nusrat
A
,
Parkos
CA
(
2006
).
Microtubules regulate disassembly of epithelial apical junctions
.
BMC Cell Biol
7
,
12
.
Jarrett
CR
,
Blancato
J
,
Cao
T
,
Bressette
DS
,
Cepeda
M
,
Young
PE
,
King
CR
,
Byers
SW
(
2001
).
Human APC2 localization and allelic imbalance
.
Cancer Res
.
61
,
7978
7984
.
Ji
H
,
Ramsey
MR
,
Hayes
DN
,
Fan
C
,
McNamara
K
,
Kozlowski
P
,
Torrice
C
,
Wu
MC
,
Shimamura
T
,
Perera
SA
, et al.
(
2007
).
LKB1 modulates lung cancer differentiation and metastasis
.
Nature
448
,
807
810
.
Jimbo
T
,
Kawasaki
Y
,
Koyama
R
,
Sato
R
,
Takada
S
,
Haraguchi
K
,
Akiyama
T
(
2002
).
Identification of a link between the tumour suppressor APC and the kinesin superfamily
.
Nat Cell Biol
.
4
,
323
327
.
Joukov
V
,
Groen
AC
,
Prokhorova
T
,
Gerson
R
,
White
E
,
Rodriguez
A
,
Walter
JC
,
Livingston
DM
(
2006
).
The BRCA1/BARD1 heterodimer modulates ran-dependent mitotic spindle assembly
.
Cell
127
,
539
552
.
Kaelin
WG
Jr
(
2007
).
The von hippel-lindau tumor suppressor protein and clear cell renal carcinoma
.
Clin Cancer Res
.
13
,
680s
684s
.
Kaelin
WG
Jr
(
2008
).
The von hippel-lindau tumour suppressor protein: O2 sensing and cancer
.
Nat Rev Cancer
8
,
865
873
.
Kamada
M
,
Suzuki
K
,
Kato
Y
,
Okuda
H
,
Shuin
T
(
2001
).
von Hippel-Lindau protein promotes the assembly of actin and vinculin and inhibits cell motility
.
Cancer Res
.
61
,
4184
4189
.
Kaplan
KB
,
Burds
AA
,
Swedlow
JR
,
Bekir
SS
,
Sorger
PK
,
Nathke
IS
(
2001
).
A role for the adenomatous polyposis coli protein in chromosome segregation
.
Nat Cell Biol
.
3
,
429
432
.
Kearney
TJ
,
Price
EA
,
Lee
S
,
Silberman
AW
(
1993
).
Tumor aneuploidy in young patients with colorectal cancer
.
Cancer
72
,
42
45
.
Klymkowsky
MW
,
Savagner
P
(
2009
).
Epithelial-mesenchymal transition: a cancer researcher’s conceptual friend and foe
.
Am J Pathol
.
174
,
1588
1593
.
Kojima
Y
,
Miyoshi
H
,
Clevers
HC
,
Oshima
M
,
Aoki
M
,
Taketo
MM
(
2007
).
Suppression of tubulin polymerization by the LKB1-Microtubule-associated protein/microtubule affinity-regulating kinase signaling
.
J Biol Chem
.
282
,
23532
23540
.
Kroboth
K
,
Newton
IP
,
Kita
K
,
Dikovskaya
D
,
Zumbrunn
J
,
Waterman-Storer
CM
,
Näthke
IS
(
2007
).
Lack of adenomatous polyposis coli protein correlates with a decrease in cell migration and overall changes in microtubule stability
.
Mol Biol Cell
18
,
910
918
.
Kuehn
EW
,
Walz
G
,
Benzing
T
(
2007
).
Von hippel-lindau: a tumor suppressor links microtubules to ciliogenesis and cancer development
.
Cancer Res
.
67
,
4537
4540
.
Lallemand
D
,
Curto
M
,
Saotome
I
(
2003
).
NF2 deficiency promotes tumorigenesis and metastasis by destabilizing adherens junctions
.
Genes Dev
.
17
,
1090
1100
.
Lee
JH
,
Koh
H
,
Kim
M
,
Kim
Y
,
Lee
SY
,
Karess
RE
,
Lee
S-H
,
Shong
M
,
Kim
J-M
,
Kim
J
, et al.
(
2007
).
Energy-dependent regulation of cell structure by AMP-activated protein kinase
.
Nature
477
,
1017
1021
.
Lerwill
MF
(
2004
).
Current practical applications of diagnostic immunohistochemistry in breast pathology
.
Am J Surg Pathol
.
28
,
1076
1091
.
Ligon
LA
,
Holzbaur
EL
(
2007
).
Microtubules tethered at epithelial cell junctions by dynein facilitate efficient junction assembly
.
Traffic
8
,
808
819
.
Lingle
WL
,
Lutz
WH
,
Ingle
JN
,
Haihle
NJ
,
Salisbury
JL
(
1998
).
Centrosome hypertrophy in human breast tumors: implications for genomic stability and cell polarity
.
Proc Natl Acad Sci USA
95
,
2950
2955
.
Liu
L
,
Tommasi
S
,
Lee
D-H
,
Dammann
R
,
Pfeifer
GP
(
2003
).
Control of microtubule stability by the RASSF1A tumor suppressor
.
Oncogene
22
,
8125
8136
.
Liu
L
,
Guo
C
,
Dammann
R
,
Tommasi
S
,
Pfeifer
GP
(
2008
).
RASSF1A interacts with and activates the mitotic kinase Aurora-A
.
Oncogene
27
,
6175
6186
.
Lolkema
MP
,
Mehra
N
,
Jorna
AS
,
van Beest
M
,
Giles
RH
,
Voest
EE
(
2004
).
The von Hippel-Lindau tumor suppressor protein influences microtubule dynamics at the cell periphery
.
Exp Cell Res
.
301
,
139
146
.
Lolkema
MP
,
Mans
DA
,
Snijckers
CM
,
van Noort
M
,
van Beest
M
,
Voest
EE
,
Giles
RH
(
2007
).
The von Hippel-Lindau tumour suppressor interacts with microtubules through kinesin-2
.
FEBS Lett
.
581
,
4571
4576
.
Louie
RK
,
Bahmanyar
S
,
Siemers
KA
,
Votin
V
,
Chang
P
,
Stearns
T
,
Nelson
WJ
,
Barth
AIM
(
2004
).
Adenomatous polyposis coli and EB1 localize in close proximity of the mother centriole and EB1 is a functional component of centrosomes
.
J Cell Sci
.
117
,
1117
1128
.
Lutz
MS
,
Burk
RD
(
2006
).
Primary cilium formation requires von hippel-lindau gene function in renal-derived cells
.
Cancer Res
.
66
,
6903
6907
.
Malkin
D
(
1994
).
Germline p53 mutations and heritable cancer
.
Annu Rev Genet
.
28
,
443
465
.
Manavathi
B
,
Acconcia
F
,
Rayala
SK
,
Kumar
R
(
2006
).
An inherent role of microtubule network in the action of nuclear receptor
.
Proc Natl Acad Sci USA
103
,
15981
15986
.
Mani
SA
,
Guo
W
,
Liao
MJ
,
Eaton
EN
,
Ayyanan
A
,
Zhou
AY
,
Brooks
M
,
Reinhard
F
,
Zhang
CC
,
Shipitsin
M
, et al.
(
2008
).
The epithelial-mesenchymal transition generates cells with properties of stem cells
.
Cell
133
,
704
715
.
Masciari
S
,
Larsson
N
,
Senz
J
,
Boyd
N
,
Kaurah
P
,
Kandel
MJ
,
Harris
LN
,
Pinheiro
HC
,
Troussard
A
,
Miron
P
, et al.
(
2007
).
Germline E-cadherin mutations in familial lobular breast cancer
.
J Med Genet
.
44
,
726
731
.
Massague
J
,
Weinberg
RA
(
1992
).
Negative regulators of growth
.
Curr Opin Genet Dev
.
2
,
28
32
.
Matsumoto
S
,
Iwakawa
R
,
Takahashi
K
,
Kohno
T
,
Nakanishi
Y
,
Matsuno
Y
,
Suzuki
K
,
Nakamoto
M
,
Shimizu
E
,
Minna
JD
, et al.
(
2007
).
Prevalence and specificity of LKB1 genetic alterations in lung cancers
.
Oncogene
26
,
5911
5918
.
McClatchey
AI
,
Fehon
RG
(
2009
).
Merlin and the ERM proteins-regulators of receptor distribution and signaling at the cell cortex
.
Trends Cell Biol
.
19
,
198
206
.
Meng
W
,
Mushika
Y
,
Ichii
T
,
Takeichi
M
(
2008
).
Anchorage of microtubule minus ends to adherens junctions regulates epithelial cell-cell contacts
.
Cell
135
,
948
959
.
Mimori-Kiyosue
Y
,
Shiina
N
,
Tsukita
S
(
2000
).
Adenomatous polyposis coli (APC) protein moves along microtubules and concentrates at their growing ends in epithelial cells
.
J Cell Biol
.
148
,
505
517
.
Mimori-Kiyosue
Y
,
Matsui
C
,
Sasaki
H
,
Tsukita
S
(
2007
).
Adenomatous polyposis coli (APC) protein regulates epithelial cell migration and morphogenesis via PDZ domain-based interactions with plasma membranes
.
Genes Cells
12
,
219
233
.
Mitchison
TJ
(
1986
).
Beyond self-assembly: from microtubules to morphogenesis
.
Cell
45
,
329
342
.
Morris
PG
,
Fornier
MN
(
2008
).
Microtubule active agents: beyond the taxane frontier
.
Clin Cancer Res
.
14
,
7167
7172
.
Morris
VB
,
Brammall
J
,
Noble
J
,
Reddel
R
(
2000
).
p53 Localizes to the centrosomes and spindles of mitotic cells in the embryonic chick epiblast, human cell lines, and a human primary culture: an immunofluorescence study
.
Exp Cell Res
.
256
,
122
130
.
Morrison
SJ
,
Kimble
J
(
2006
).
Asymmetric and symmetric stem-cell divisions in development and cancer
.
Nature
441
,
1068
1074
.
Munemitsu
S
,
Souza
B
,
Muller
O
,
Albert
I
,
Bubinfeld
B
,
Polakis
P
(
1994
).
The APC gene product associates with microtubules in vivo and promotes their assembly in vitro
.
Cancer Res
54
,
3676
3681
.
Muranen
T
,
Gronholm
M
,
Lampin
A
,
Lallemand
D
,
Zhao
F
,
Giovannini
M
,
Carpen
O
(
2007
).
The tumor suppressor merlin interacts with microtubules and modulates Schwann cell microtubule cytoskeleton
.
Hum Mol Genet
.
16
,
1742
1751
.
Musch
A
(
2004
).
Microtubule organization and function in epithelial cells
.
Traffic
5
,
1
9
.
Nakamura
M
,
Zhou
XZ
,
Lu
KP
(
2001
).
Critical role for the EB1 and APC interaction in the regulation of microtubule polymerization
.
Curr Biol
.
11
,
1062
1067
.
Nathke
IS
(
2000
).
The adenomatous polyposis coli protein
.
Mol Pathol
.
52
,
169
173
.
Nathke
IS
(
2004
).
The adenomatous polyposis coli protein: the Achilles heel of the gut epithelium
.
Annu Rev Cell Biol
.
20
,
337
366
.
Nathke
IS
(
2005
).
Relationship between the role of the adenomatous polyposis coli protein in colon cancer and its contribution to cytoskeletal regulation
.
Biochem Soc Trans
.
33
,
694
697
.
Nathke
IS
,
Adams
CL
,
Polakis
P
,
Sellin
JH
,
Nelson
WJ
(
1996
).
The adenomatous polyposis coli tumor suppressor protein localizes to plasma membrane sites involved in active cell migration
.
J Cell Biol
.
134
,
165
179
.
Nishisho
I
,
Nakamura
Y
,
Miyoshi
Y
,
Miki
Y
,
Ando
H
,
Horii
A
,
Koyama
K
,
Utsunomiya
J
,
Baba
S
,
Hedge
P
(
1991
).
Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients
.
Science
253
,
665
669
.
Nowak
MA
,
Komarova
NL
,
Sengupta
A
,
Jallepalli
PV
,
Shih
l-M
,
Vogelstein
B
,
Lengauer
C
(
2002
).
The role of chromosomal instability in tumor initiation
.
Proc Natl Acad Sci USA
99
,
16226
16231
.
Nyhan
MJ
,
O’Sullivan
GC
,
McKenna
SL
(
2008
).
Role of the VHL (von Hippel-Lindau) gene in renal cancer: a multifunctional tumour suppressor
.
Biochem Soc Trans
.
36
,
472
478
.
Orth
JD
,
Tang
Y
,
Shi
J
,
Loy
CT
,
Amendt
C
,
Wilm
C
,
Zenke
FT
,
Mitchison
TJ
(
2008
).
Quantitative live imaging of cancer and normal cells treated with Kinesin-5 inhibitors indicates significant differences in phenotypic responses and cell fate
.
Mol Cancer Ther
.
7
,
3480
3489
.
Ortiz-Vega
S
,
Khokhlatchev
A
,
Nedwidek
M
,
Zhang
XF
,
Dammann
R
,
Pfeifer
GP
,
Avruch
J
(
2002
).
The putative tumor suppressor RASSF1A homodimerizes and heterodimerizes with the Ras-GTP binding protein Nore1
.
Oncogene
21
,
1381
1390
.
Pellman
D
(
2007
).
Aneuploidy and cancer
.
Nature
446
,
38
39
.
Perez-Moreno
M
,
Jamora
C
,
Fuchs
E
(
2003
).
Sticky business: orchestrating cellular signals at adherens junctions
.
Cell
112
,
535
548
.
Pihan
GA
,
Wallace
J
,
Zhou
Y
,
Doxsey
SJ
(
2003
).
Centrosome abnormalities and chromosome instability occur together in pre-invasive carcinomas
.
Cancer Res
.
63
,
1398
1404
.
Polyak
K
,
Weinberg
RA
(
2009
).
Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits
.
Nat Rev Cancer
9
,
265
273
.
Reed
N
,
Gutmann
DH
(
2001
).
Tumorigenesis in neurofibromatosis: new insights and potential therapies
.
Trends Mol Med
.
7
,
157
162
.
Resta
N
,
Stella
A
,
Susca
F
,
Montera
M
,
Gentile
M
,
Cariola
F
,
Prete
F
,
Tenconi
R
,
Tibiletti
MG
,
Logrieco
G
, et al.
(
2001
).
Nine novel APC mutations in Italian FAP patients
.
Hum Mutat
.
17
,
434
435
.
Richter
AM
,
Pfeifer
GP
,
Dammann
RH
(
2009
).
The RASSF proteins in cancer; from epigenetic silencing to functional characterization
.
Biochemica Biophysica Acta
1796
,
114
128
.
Rong
R
,
Jin
W
,
Zhang
J
,
Sheikh
MS
,
Huang
Y
(
2004
).
Tumor suppressor RASSF1A is a microtubule-binding protein that stabilizes microtubules and induces G2/M arrest
.
Oncogene
23
,
8216
8230
.
Rong
R
,
Jiang
LY
,
Sheikh
MS
,
Huang
Y
(
2007
).
Mitotic kinase Aurora-A phosphorylates RASSF1A and modulates RASSF1A-mediated microtubule interaction and M-phase cell cycle regulation
.
Oncogene
26
,
7700
7708
.
Rubinfeld
B
,
Albert
I
,
Porfiri
E
,
Fiol
C
,
Menumitsu
S
,
Polakis
P
(
1996
).
Binding of GSK3β to the APC-βCatenin complex and regulation of complex assembly
.
Science
272
,
1023
1026
.
Rusan
NM
,
Peifer
M
(
2008
).
Original CIN: reviewing roles for APC in chromosome instability
.
J Cell Biol
.
181
,
719
726
.
Sankaran
S
,
Crone
DE
,
Palazzo
RE
,
Parvin
JD
(
2007
).
BRCA1 Regulates γ-tubulin binding to centrosomes
.
Cancer Biol Ther
.
6
,
1853
1857
.
Sansom
OJ
,
Reed
KR
,
Hayes
AJ
,
Ireland
H
,
Brinkmann
H
,
Newton
IP
,
Batlle
E
,
Simon-Assmann
P
,
Clevers
H
,
Nathke
IS
, et al.
(
2004
).
Loss of Apc in vivo immediately perturbs Wnt signaling, differentiation, and migration
.
Genes Dev
.
18
,
1385
1390
.
Schermer
B
,
Ghenoiu
C
,
Malte Müller
RU
,
Kotsis
F
,
Höhne
M
,
Kühn
W
,
Rapka
M
,
Nitschke
R
,
Zentgraf
H
,
Fliegauf
M
, et al.
(
2006
).
The von Hippel-Lindau tumor suppressor protein controls ciliogenesis by orienting microtubule growth
.
J Cell Biol
.
175
,
547
554
.
Schmalhofer
O
,
Brabletz
S
,
Brabletz
T
(
2009
).
E-cadherin, β-catenin, and ZEB1 in malignant progression of cancer
.
Cancer Metastasis Rev
.
28
,
151
166
.
Scoles
DR
(
2008
).
The merlin interacting proteins reveal multiple targets for NF2 therapy
.
Biochem Biophys Acta
1785
,
32
54
.
Shaw
RM
,
Fay
AJ
,
Puthenveedu
MA
,
von Zastrow
M
,
Jan
YN
,
Jan
LY
(
2007
).
Microtubule plus-end-tracking proteins target gap junctions directly from the cell interior to adherens junctions
.
Cell
128
,
547
560
.
Shibata
T
,
Gotoh
M
,
Ochiai
A
,
Hirohashi
S
(
1994
).
Association of plakoglobin with APC, a tumor suppressor gene product, and its regulation by tyrosine phosphorylation
.
Biochem Biophys Res Commun
.
203
,
519
522
.
Shinmura
K
,
Bennett
RA
,
Tarapore
P
,
Fukasawa
K
(
2007
).
Direct evidence for the role of centrosomally localized p53 in the regulation of centrosome duplication
.
Oncogene
26
,
2939
2944
.
Sieber
OM
,
Tomlinson
IP
,
Lamlum
H
(
2001
).
The adenomatous polyposis coli (APC) tumour suppressor – genetics, function and disease
.
Mol Med Today
6
,
462
469
.
Smith
KJ
,
Levy
DB
,
Maupin
P
,
Pollard
TD
,
Vogelstein
B
,
Kinzler
Kw
(
1994
).
Wild-Type but not mutant APC associates with the microtubule cytoskeleton
.
Cancer Res
.
54
,
3672
3675
.
Smits
R
,
Kielman
MF
,
Breukel
C
,
Zurcher
C
,
Neufeld
K
,
Jagmohan-Changur
S
,
Hofland
N
,
van Dijk
J
,
White
R
,
Edelmann
W
, et al.
(
1999
).
Apc1638T: a mouse model delineating critical domains of the adenomatous polyposis coli protein involved in tumorigenesis and development
.
Genes Dev
.
13
,
1309
1321
.
Song
MS
,
Song
SJ
,
Ayad
NG
,
Chang
JS
,
Lee
JH
,
Hong
HK
,
Lee
H
,
Choi
N
,
Kim
J
,
Kim
H
, et al.
(
2004
).
The tumour suppressor RASSF1A regulates mitosis by inhibiting the APC-Cdc20 complex
.
Nat Cell Biol
.
6
,
129
137
.
Song
SJ
,
Song
MS
,
Kim
SJ
,
Kim
SY
,
Kwon
SH
,
Kim
JG
,
Calvisi
DF
,
Kang
D
,
Lim
D-S
(
2009
).
Aurora A regulates prometaphase progression by inhibiting the ability of RASSF1A to suppress APC-Cdc20 activity
.
Cancer Res
.
69
,
2314
2323
.
Starita
LM
,
Machida
Y
,
Sankaran
S
,
Elias
JE
,
Griffin
K
,
Schlegel
BP
,
Gygi
SP
,
Parvin
JD
(
2004
).
BRCA1-dependent ubiquitination of gamma-tubulin regulates centrosome number
.
Mol Cell Biol
.
24
,
8457
8466
.
Stehbens
SJ
,
Akhmanova
A
,
Yap
AS
(
2009
).
Microtubules and cadherins: a neglected partnership
.
Front Biosci
.
14
,
3159
3167
.
Strathdee
G
(
2002
).
Epigenetic versus genetic alterations in the inactivation of E-cadherin
.
Semin Cancer Biol
.
12
,
373
379
.
Su
L-K
,
Burrell
M
,
Hill
DE
,
Gyuris
J
,
Brent
R
,
Wiltshire
R
,
Trent
J
,
Vogelstein
B
,
Kinzler
KW
(
1995
).
APC binds to the novel protein EB1
.
Cancer Res
.
55
,
2972
2977
.
Szczepanska
K
,
Maleszewski
M
(
2005
).
LKB1/PAR4 protein is asymmetrically localized in mouse oocytes and associates with meiotic spindle
.
Gene Expr Patterns
6
,
86
93
.
Tarapore
P
,
Tokuyama
Y
,
Horn
HF
,
Fukasawa
K
(
2001
).
Difference in the centrosome duplication regulatory activity among p53 ‘hot spot’ mutants: potential role of Ser 315 phosphorylation-dependent centrosome binding of p53
.
Oncogene
20
,
6851
6863
.
Tetsu
O
,
McCormick
F
(
1999
).
Beta-catenin regulates expression of Cyclin D1 in colon carcinoma cells
.
Nature
398
,
422
426
.
Thoma
CR
,
Frew
IJ
,
Hoerner
CR
,
Montani
M
,
Moch
H
,
Krek
W
(
2007
).
pVHL and GSK3beta are components of a primary cilium-maintenance signalling network
.
Nat Cell Biol
.
9
,
588
595
.
Tighe
A
,
Johnson
VL
,
Albertella
M
,
Taylor
SS
(
2001
).
Aneuploid colon cancer cells have a robust spindle checkpoint
.
EMBO Rep
.
2
,
609
614
.
Tommasi
S
,
Dammann
R
,
Zhang
Z
,
Wang
Y
,
Liu
L
,
Tsark
WM
,
Wilczynski
SP
,
Li
J
,
You
M
,
Pfeifer
GP
(
2005
).
Tumor susceptibility of Rassf1a knockout mice
.
Cancer Res
.
65
,
92
98
.
Tritarelli
A
,
Oricchio
E
,
Ciciarello
M
,
Mangiacasale
R
,
Palena
A
,
Lavia
P
,
Soddu
S
,
Cundari
E
(
2004
).
p53 localization at centrosomes during mitosis and postmitotic checkpoint are ATM-dependent and require serine 15 phosphorylation
.
Mol Biol Cell
15
,
8
.
Trovo-Marqui
AB
,
Tajara
EH
(
2006
).
Neurofibromin: a general outlook
.
Clin Genet
.
70
,
1
13
.
Tsukita
S
,
Tsukita
S
,
Nagafuchi
A
,
Yonemura
S
(
1992
).
Molecular linkage between cadherins and actin filaments in cell-cell adherens junctions
.
Curr Opin Cell Biol
.
4
,
834
839
.
van der Weyden
L
,
Tachibana
KK
,
Gonzalez
MA
,
Adams
DJ
,
Ng
BL
,
Petty
R
,
Venkitaraman
AR
,
Arends
MJ
,
Bradley
A
(
2005
).
The RASSF1A isoform of RASSF1 promotes microtubule stability and suppresses tumorigenesis
.
Mol Cell Biol
.
25
,
8356
8367
.
van Es
JH
,
Kirkpatrick
C
,
van de Wetering
M
,
Molenaar
M
,
Miles
A
,
Kuipers
J
,
Destrée
O
,
Peifer
M
,
Clevers
H
(
1999
).
Identification of APC2, a homologue of the adenomatous polyposis coli tumour suppressor
.
Curr Biol
.
9
,
105
108
.
van Roy
F
,
Berx
G
(
2008
).
The cell-cell adhesion molecule E-cadherin
.
Cell Mol Life Sci
.
65
,
3756
3788
.
Vos
MD
,
Martinez
A
,
Elam
C
,
Dallol
A
,
Taylor
BJ
,
Latif
F
,
Clark
GJ
(
2004
).
A Role for the RASSF1A tumor suppressor in the regulation of tubulin polymerization and genomic stability
.
Cancer Res
.
64
,
4244
4250
.
Wadsworth
P
,
Bottaro
DP
(
1996
).
Microtubule dynamic turnover is suppressed during polarization and stimulated in hepatocyte growth factor scattered madindarby canine kidney epthelial cells
.
Cell Motil Cytoskeleton
35
,
225
236
.
Watanabe
T
,
Wang
S
,
Noritake
J
,
Sato
K
,
Fukata
M
,
Takefuji
M
,
Nakagawa
M
,
Izumi
N
,
Akiyama
T
,
Kaibuchi
K
(
2004
).
Interaction with IQGAP1 links APC to Rac1, Cdc42 and actin filaments during cell polarization and migration
.
Dev Cell
7
,
871
873
.
Waterman-Storer
CM
,
Salmon
WC
,
Salmon
ED
(
2000
).
Feedback interactions between cell-cell adherens junctions and cytoskeletal dynamics in newt lung epithelial cells
.
Mol Biol Cell
11
,
2471
2483
.
Watts
JL
,
Morton
DG
,
Bestman
J
,
Kemphues
KJ
(
2000
).
The C. elegans par-4 gene encodes a putative serine-threonine kinase required for establishing embryonic asymmetry
.
Development
127
,
1467
1475
.
Wen
Y
,
Eng
CH
,
Schmoranzer
J
,
Cabrera-Poch
N
,
Morris
EJS
,
Chen
M
,
Wallar
BJ
,
Alberts
AS
,
Gundersen
GG
(
2004
).
EB1 and APC bind to mDia to stabilize microtubules downstream of Rho and promote cell migration
.
Nat Cell Biol
.
6
,
820
830
.
Wong
M
,
Hermiston
ML
,
Syder
AJ
,
Gordon
JI
(
1996
).
Forced expression of the tumor suppressor adenomatous polyposis coli protein induces disordered cell migration in the intestinal epithelium
.
Proc Natl Acad Sci USA
93
,
9588
9593
.
Wong
NA
,
Pignatelli
M
(
2002
).
Beta-catenin-a linchpin in colorectal carcinogenesis?
Am J Pathol
.
160
,
389
401
.
Xiao
G-H
,
Chernoff
J
,
Testa
JR
(
2003
).
NF2: The Wizardry of Merlin
.
Genes Chromosomes Cancer
38
,
389
399
.
Xu
H-M
,
Gutmann
DH
(
1998
).
Merlin differentially associates with the microtubule and actin cytoskeleton
.
J Neurosci Res
.
51
,
403
415
.
Xu
X
,
Weaver
Z
,
Linke
SP
,
Li
C
,
Gotay
J
,
Wang
XW
,
Harris
CC
,
Ried
T
,
Deng
CX
(
1999
).
Centrosome amplification and a defective G2-M cell cycle checkpoint induce genetic instability in BRCA1 exon 11 isoform-deficient cells
.
Mol Cell
3
,
389
395
.
Yap
AS
,
Stevenson
BR
,
Abel
KC
,
Cragoe
EJJ
,
Manley
SW
(
1995
).
Microtubule integrity is necessary for the epithelial barrier function of cultured thyroid cell monolayers
.
Exp Cell Res
.
218
,
540
550
.
Yu
W
,
O’Brien
LE
,
Wang
F
,
Bourne
H
,
Mostov
KE
,
Zegers
MM
(
2003
).
Hepatocyte growth factor switches orientation of polarity and mode of movement during morphogenesis of multicellular epithelial structures
.
Mol Biol Cell
14
,
748
763
.
Yu
X
,
Waltzer
L
,
Bienz
M
(
1999
).
A new Drosophila APC homologue associated with adhesive zones of epithelial cells
.
Nat Cell Biol
.
1
,
144
151
.
Zhang
J
,
Ahmad
S
,
Mao
Y
(
2007
).
BubR1 and APC/EB1 cooperate to maintain metaphase chromosome alignment
.
J Cell Biol
.
178
,
773
784
.
Zhang
J
,
Neisa
R
,
Mao
Y
(
2009
).
Oncogenic Adenomatous polyposis coli mutants impair the mitotic checkpoint through direct interaction with Mad2
.
Mol Biol Cell
20
,
2381
2388
.
Zhang
S
,
Schafer-Hales
K
,
Khuri
FR
,
Zhou
W
,
Vertino
PM
,
Marcus
AI
(
2008
).
The tumor suppressor LKB1 regulates lung cancer cell polarity by mediating cdc42 recruitment and activity
.
Cancer Res
.
68
,
740
748
.
Zumbrunn
J
,
Kinoshita
K
,
Hyman
AA
,
Nathke
IS
(
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
.
Zyss
D
,
Gergely
F
(
2009
).
Centrosome function in cancer: guilty or innocent?
Trends Cell Biol
.
19
,
334
346
.

COMPETING INTERESTS

The authors declare no competing financial interests.