Skip to main content

REVIEW article

Front. Oncol., 13 February 2023
Sec. Molecular and Cellular Oncology
This article is part of the Research Topic Molecular Regulation of Tumor Cells Migration and Metastatic Growth View all 11 articles

The emerging role of microtubules in invasion plasticity

Anna Legtov,Anna Legátová1,2Markta Pelantov,Markéta Pelantová1,2Daniel Rsel,Daniel Rösel1,2Jan Brbek,Jan Brábek1,2Aneta &#x;karkov,*Aneta Škarková1,2*
  • 1Department of Cell Biology, Charles University, Prague, Czechia
  • 2Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia

The ability of cells to switch between different invasive modes during metastasis, also known as invasion plasticity, is an important characteristic of tumor cells that makes them able to resist treatment targeted to a particular invasion mode. Due to the rapid changes in cell morphology during the transition between mesenchymal and amoeboid invasion, it is evident that this process requires remodeling of the cytoskeleton. Although the role of the actin cytoskeleton in cell invasion and plasticity is already quite well described, the contribution of microtubules is not yet fully clarified. It is not easy to infer whether destabilization of microtubules leads to higher invasiveness or the opposite since the complex microtubular network acts differently in diverse invasive modes. While mesenchymal migration typically requires microtubules at the leading edge of migrating cells to stabilize protrusions and form adhesive structures, amoeboid invasion is possible even in the absence of long, stable microtubules, albeit there are also cases of amoeboid cells where microtubules contribute to effective migration. Moreover, complex crosstalk of microtubules with other cytoskeletal networks participates in invasion regulation. Altogether, microtubules play an important role in tumor cell plasticity and can be therefore targeted to affect not only cell proliferation but also invasive properties of migrating cells.

1 Introduction

Cells have adopted various migration/invasion modes which vary in their nature of force generation and dependency on cell-cell and cell-extracellular matrix (ECM) adhesion. Collective migration requires both cell-cell and cell-ECM adhesion, mesenchymal migration omits intercellular adhesion but strongly relies on cell-ECM contact and amoeboid migration can be independent of adhesion altogether. The large range of invasion modes ensures physiological migration of cells in various environments, but in the hands of cancer cells it has become a dangerous trait. The ability of cancer cells to utilize one or more of the invasion modes, and to switch among them in response to changing circumstances is termed invasion plasticity and represents a large complication on the road to treating metastatic disease.

Due to the requirements for dynamic changes of cell morphology during invasion, it is evident the invading cell must readily reorganize its cytoskeleton (14). This is even more prominent in cells with high invasion plasticity that switch among the elongated mesenchymal and round amoeboid phenotype, in a process termed mesenchymal-amoeboid transition (MAT) or amoeboid-mesenchymal transition (AMT) (57). During MAT, cells retract protrusions, round up and initiate intense membrane blebbing, which may be due to loss in cell adhesivity (8) and/or fast increase of hydrostatic pressure that detaches the membrane from the cortex (9). The rapid membrane blebbing is often reduced after transitioning to a motile amoeboid phenotype. Opposingly, AMT is accompanied by loss of blebbing activity and cell elongation through stabilization of protrusions.

Actin reorganization in migrating cells is well described, with RhoGTPases playing a key role (10). Rac and Cdc42 are known to be responsible for promoting actin polymerization leading to the formation of lamellipodia and filopodia as a result of stimulating the Arp2/3 complex through activation of either WASP or SCARE/WAVE family (1113). Due to its function as an initiator of lamellipodia formation, Rac is preferentially active at the leading edge of migrating cells (14). Opposingly, RhoA activity is higher at the cell rear, where its signaling mediates rear contractility and detachment (15). This is achieved by RhoA-mediated activation of ROCK, which in turn leads to phosphorylation (therefore inhibition) of MLCP, resulting in higher phosphorylation of the myosin light chain and increased contractility (16, 17). ROCK is also responsible for the phosphorylation of LIMK and subsequently of cofilin, which stabilizes actin bundles (18), resulting in formation of stress fibers. Due to the different requirement of protrusive activity and contractility of mesenchymal and amoeboid migration, each is dominated by different RhoGTPase signaling. Amoeboid cells require RhoA/ROCK signaling for their migration and inhibition of this pathway leads to a switch to mesenchymal invasion (19, 20). Similarly, inducing constitutively active RhoA/ROCK can induce the mesenchymal-to-amoeboid switch (21, 22). On the other hand, Rac signaling promotes mesenchymal traits (23, 24).

Moreover, signaling mediated by RhoGTPases interconnects the actin cytoskeleton with the microtubule (MT) network (Figure 1). For example, in fibroblasts, MTs growth stimulates Rac1 activity, therefore promoting lamellipodia formation (25, 26). On the other hand, Rac1/Cdc42 signaling can lead to MTs polymerization via stathmin inhibition (27, 28), and RhoA can promote stabilization of MTs by its effector, mDia1, which interacts with MTs and induces their capping and alignment with actin bundles (29, 30).

FIGURE 1
www.frontiersin.org

Figure 1 Microtubules and RhoGTPase signaling. (A) The length and stability of MTs in migrating cells is interconnected with RhoGTPase signaling and depends on cell polarization. MTs preferentially elongate toward the leading edge, where their growth is enhanced by various MT end-binding proteins and MT stabilizing proteins, which further interact with numerous Rac1 and Cdc42 activators (see TRIO, TIAM and complex IQGAP/CLIP-170). At the leading edge, Rac1 and Cdc42 signaling contributes to actin polymerization (pink network), but also MT stabilization by phosphorylation of stathmin. MT stability at the leading edge is supported by p27Kip, which binds stathmin, preventing its activation. Similarly, p27Kip prevents RhoA pathway activation. On the contrary, at the trailing edge, MTs are depolymerized and Rho/ROCK signaling pathway dominates. Stathmin is not phosphorylated by Rac1 or Cdc42 and remains active – able to sequester tubulin dimers and destabilize MTs. MT disruption leads to release, and thus activation, of GEFH1 from MTs into the cytoplasm, where it promotes activation of the RhoA/ROCK pathway. Roman numerals labeling MTs refer to part (B) of the Figure. (B) Schematic illustration showing interaction between MT stabilizing (green)/destabilizing (red) factors and RhoGTPases. Created with BioRender.com.

Apart from actin, MTs also interact with intermediate filaments (IFs). This can be either indirectly through linker proteins, such as APC, or directly, and the mutual interaction stabilizes MTs and promotes directed migration (31, 32).

The role of microtubules in cell migration is multifaceted, encompassing intracellular transport and delivery of migration associated cargo, protrusion stabilization and regulation of adhesions (3335). Less is known about the role of microtubules in 3D migration, yet alone specifically in amoeboid or mesenchymal invasion. Thus, we would like to summarize current knowledge on the role of microtubule cytoskeleton in cell invasion in the context of mesenchymal and amoeboid phenotypes and transitions among them (Figure 2).

FIGURE 2
www.frontiersin.org

Figure 2 Microtubules in different invasive modes. In mesenchymal cells (left), MTs are elongated and stabilized at the front and depolymerized at the retracting end. Mesenchymal cells contain many adhesion structures, such as focal adhesions and invadopodia, that are tightly coupled to MT dynamics. At the leading edge, protrusive activity is regulated by CLASP and SLAIN, which reduce MT catastrophes and contribute to MT growth persistence and elongation of protrusions. In mesenchymal cells, GEFH1 is predominantly bound to MTs, which keeps it in an inactive state. On the other hand, amoeboid migration modes (right) are generally associated with a less stable MT network. Accordingly, increased stathmin activity or downregulation of MT-stabilizing proteins such as mDia2 or p27Kip has been shown to cause the mesenchymal-amoeboid transition (MAT). In pseudopodal amoeboid cells, MTs contribute to circumnavigation and pseudopod extension. In blebby amoeboid cells, MTs are generally disrupted and thus MT-destabilizing drugs, such as vincristine and nocodazole, promote transition to the blebby amoeboid phenotype. In amoeboid cells, GEFH1 can be found free in the cytoplasm, where it activates the RhoA/ROCK pathway. Note that the MTOC is located in front of the nucleus in both mesenchymal and amoeboid cells, although in some pseudopodal amoeboid cells, such as leukocytes, the MTOC is found at the cell rear. This information, together with the basic characteristics of the individual invasion modes, is summarized in the table at the bottom of the figure. Created with BioRender.com.

2 Microtubule associated proteins in cell migration

The stability and dynamics of the MT network are modulated by numerous MT associated proteins, some of which have been directly linked to cell invasion plasticity, see below and in Figure 1.

2.1 EB1

End binding protein 1 (EB1) binds +end of MTs, localizing to the distal tips of MTs, the centrosome or MT ends in the mitotic spindle. It is sometimes referred to as the „master regulator of plus-end-tracking proteins (+TIPs)” for its ability to recruit various +TIPs and thus influence not only MTs themselves, but other processes such as membrane-anchoring or actin polymerization as well (36). EB1 binding to MTs stimulates MT elongation, whereas its dissociation causes slower growth of MTs and can influence cells’ direction of movement (37). Interestingly, depletion of EB1 has much bigger consequences on migration and protrusion branching in 3D migration. Whereas in 2D there is no significant effect on protrusions and overall migration speed, in 3D, depleting EB1 in mesenchymally migrating cells results in slower invasion and defects in cell directionality (38). In mesenchymal cells, EB1 mediates the binding of proteins CLASP1 and SLAIN2 to MTs, which contributes to their growth persistence in protrusions by reducing catastrophes (Figure 2). This was shown to be necessary for the invasive shape and 3D mesenchymal invasion (39).

2.2 IQGAP

IQ motif-containing GTPase-activating protein 1/2/3 (IQGAP1-3) are scaffold proteins that integrate many signaling pathways via direct and indirect binding of over 90 proteins (40), including RhoGTPases (Figure 1).

IQGAP proteins directly affects the dynamics of both the actin and the microtubule cytoskeleton. IQGAP1 is able to cross-link F-actin filaments (4144) or by binding barbed ends of actin filaments, inhibit their growth, and protect them from depolymerization (42). IQGAP1 interacts with N-WASP and Arp2/3 forming a complex able to nucleate branched actin filaments (45, 46). In agreement, IQGAP1 was shown to be localized at the leading edges of polarized cells and in the lamellipodia of motile cells (43, 44, 4648). One of the main functions of IQGAP1 is anchoring MTs to the cell cortex enabling directional movement.

With regard to MTs in migration, one of the main functions of IQGAP1 is anchoring MTs to the cell cortex enabling directional movement, a process regulated by multiple mechanisms. IQGAP1 interacts with +TIP CLIP-170 and activated Rac1 and Cdc42 (but not RhoA) forming a tripartite complex leading MTs to the cortex to areas with activated Rac1/Cdc42. Impaired binding of IQGAP1 caused by mutation at its C-terminus results in multiple leading edges in cells (49, 50). In addition, IQGAP1 and active Rac1/Cdc42 form a complex with adenomatous polyposis coli (APC) cortical filaments and depletion of either APC or IQGAP1 inhibits polarized migration (51). Another +TIP linking IQGAP1 to MTs at the cell cortex is protein SKAP (+TIP known to bind to EB1), yet again, disrupting the interaction of SKAP and IQGAP1 impairs cell migration (52).

Overall, IQGAP1 overexpression can promote cell migration and neurite outgrowth, while its depletion leads to decreased cell migration (47, 53, 54), providing evidence that the actin/microtubule crosstalk is necessary for polarized, directed cell movement.

2.3 Navigators

Navigator proteins 1-3 (NAV1/2/3) are microtubule + end binding proteins that are implicated in axon guidance and neurite outgrowth in the brain (55, 56). In neurons, NAV1 is localized at the neurite tips where it binds actin-rich domains and crosslinks with the MTs in an EB1-dependant manner (55, 57). It also forms a complex with the protein TRIO, a guanine nucleotide exchange factor (GEF) known to activate Rac1 and RhoG (5659). Thus, NAV1 does not influence MTs polymerization per se, but it promotes +end rescue and prevents catastrophes in the F-rich-domain periphery. Cells depleted in NAV1 were shown to be defective in migration during embryonic development (55). Similarly, NAV3 binds to MTs +end via EB1 and increases their polarized growth in response to EGF signaling in cancer cells by protecting them from catastrophes. Cells overexpressing NAV3 displayed higher MTs acetylation and resistance to nocodazole treatment, both distinctive of stabilized MTs (60). On the other hand, cells depleted in NAV3 showed more random migration and lost the ability to migrate persistently after EGF induction (60). Since persistent, protrusion dependent migration is characteristic of mesenchymal cells, Navigators are likely to be more crucial for mesenchymal than amoeboid migration.

2.4 Stathmin 1

Stathmin, also known as oncoprotein 18 (Opt18), is an important microtubule destabilizing protein able to cause MT depolymerization. Two modes of action have been described – first, by stimulating MT catastrophes (61), and second, by sequestrating αβ tubulin dimers to prevent their assembly (62) - which seems to depend on environmental conditions (63).

Stathmin is regulated by several kinases, and its phosphorylation on at least one of its four serins suppresses its destabilizing activity (64, 65). Moreover, Stat3 can bind to stathmin at its tubulin-binding site to prevent its function (66). Similarly, p27Kip (see further) binding prevents stathmin-regulated MT destabilization (67, 68). Of note, p27Kip expression is partially regulated by Stat3 (69).

The regulation of stathmin phosphorylation is key for cell migration, as it enables creation of a gradient of MT stability from the leading edge to the trailing edge. At the leading edge, high levels of Cdc42 and Rac1 prevent stathmin activation (27) resulting in stabilized MTs in these parts of the cell. On the trailing edge stathmin is not phosphorylated by Cdc42 or Rac and its MTs-destabilizing activity increases, leading to MT network disassembly and proper contraction of this end during cell movement (70) (Figure 1).

Generally, stathmin is a strong pro-migratory factor as evidenced by a number of studies (67, 71), although in in certain conditions its inhibition may stimulate migration (66). This may be dependent on whether the cells utilize the mesenchymal and amoeboid type of invasion, since stathmin SQ18E, which is unable to undergo inhibitory phosphorylation, was directly linked to promotion of the round, amoeboid-like phenotype in sarcoma cells (71) (Figure 2).

2.5 P27Kip

Protein p27Kip is mainly known for its nuclear role as a cyclin dependent kinase inhibitor and thus inhibitor of cell cycle progression (72). However, it also plays an important role in the cytoplasm where it interacts with other proteins through its C-terminal domain to modulate cell motility and tumor progression (67, 73, 74). One of the interaction partners of p27Kip is stathmin, binding of which inhibits stathmin´s activity leading to more stable MTs. In mesenchymal cells, this interaction limits migratory potential (67, 68) (Figure 1).

P27Kip does not influence only the MT network, it also affects actin filament reorganization. It was shown that p27Kip is able to interact with RhoA, inhibiting its ability to bind GEFs and thus preventing its activation (73). Cytoplasmatic p27Kip affects the actin cytoskeleton also indirectly via Rac1 dependent actin rearrangement and polymerization, leading to an increase in cell migration (75).

Of note, transformed fibroblasts lacking p27 Kip adopted a rounded morphology with cortical actin formation and loss of β1 integrin clusters, corresponding to the mesenchymal-amoeboid transition (76). Here both the actin and MT cytoskeleton were altered, showing a synergic effect of p27 Kip in regulation of cell invasion. In macrophages, p27 Kip contributes to the onset of mesenchymal migration by inhibition of RhoA/ROCK and lack of p27 Kip promotes amoeboid migration (77). Collectively, cytoplasmic p27 regulates invasion plasticity and exerts pro-mesenchymal signaling (Figure 2).

2.6 mDia2

mDia2 protein (mammalian homolog of Drosophila diaphanous; also known as DIAPH3) belongs to the group of formins (78), which are proteins involved in actin nucleation and elongation. In addition, mDia2 is able to bind and stabilize MT polymers. Its silencing leads to MT catastrophes and rewires EGFR and ERK signaling, resulting in increased ameboid traits (79). A different study showed that mDia2 in complex with its inhibitor induces amoeboid morphology in cells (80). Accordingly, depletion of mDia2 promoted individual dissemination of amoeboid cells from tumor spheres (81). Importantly, this affects cancer treatment, since cells without mDia2, exerting unstable MTs and amoeboid characteristics, are more susceptible to taxane chemotherapy (82). Overall, mDia2 regulates invasion plasticity through MT stabilization and actin nucleation, and its absence promotes the amoeboid invasion phenotype (Figure 2), suggesting its pro-mesenchymal role.

2.7 GEFH1

An important molecule which interconnects MT dynamics and the actomyosin network is guanosine exchange factor H1 for GTPase RhoA (GEFH1), also known as ARHGEF2. GEFH1 is a one of the few GEFs that associate with polymerized microtubules. Upon MT depolymerization, GEFH1 is released and its guanosine exchange activity increases, leading to higher activation of RhoA and its downstream signaling (Figure 1). This mechanism regulates RhoA activity in different parts of the migrating cell based on MT dynamics (83). Of note, GEFH1 activity can be further potentiated by phosphorylation mediated by Src kinase at the protruding cell edge (84). GEFH1 also affects focal adhesions (FAs) turnover in migrating cells and dysfunctional GEFH1-RhoA activation can disrupt cell motility (83). In lymphoma cells, Stat3 signaling induced destabilization of MTs, which led to the release of GEFH1 and as a result, amoeboid invasion (85).

In summary, the GEFH1-RhoA signaling pathway is induced by MTs destabilization, and its extent promotes either mesenchymal or amoeboid migration. In mesenchymal cells, activation of this pathway by dynamic growth of MTs contributes to protrusion regulation and faster FA turnover. In cells with disrupted MTs, GEFH1-RhoA signaling dominates and induces amoeboid invasion through the RhoA/ROCK pathway (Figure 2).

2.8 MAPs (MAP1B, MAP4, MAP2)

There is also a large group of microtubule associated proteins (MAPs) that bind to MTs, but surprisingly, despite their unified function of stabilizing MT, they have different roles in tumor progression and may function as both pro- and anti-metastatic factors (8688). We hypothesize that this inconsistency may be due to the different dependency of the amoeboid and mesenchymal migration on the MT network. Specifically, a change in invasive behavior was connected to MAP1B, MAP2, MAP4 and MAP7. Although there is no direct link between invasion plasticity and MAPs described so far, they likely participate by indirect signaling affecting RhoGTPases signaling. For example, MAP1B increases Rac1 activity through interaction with TIAM-1, a Rac1-GEF (89). Another possible mechanism involves phosphorylation of MAP4, which inhibits its MT stabilizing activity (90) and leads to MT disruption, which can increase RhoA through GEHH1 release (91).

2.9 Microtubule organizing center in invasion

For efficient cell migration, cells need to be polarized to maintain directionality of movement. The polarity of the cell is determined, amongst others, by the position of the nucleus and centrosome, forming the nuclear-centrosomal axis. By moving the centrosome, also known as the microtubule organizing center (MTOC), to a different position within the cell, the polarity of a cell can be shifted (92).

When cells gain a mesenchymal migratory phenotype, the centrosome position shifts to a central position in front of the nucleus relative to the future movement of the cell (93). However, this positioning of the centrosome is not a rule, as some studies show localization of the centrosome behind the nucleus (94, 95). Some studies also show that the position of the centrosome is influenced by the geometrical limitation of the cell’s surroundings rather than its function (96).

In most amoeboid cells, the location of the MTOC corresponds to mesenchymal cells. Interestingly though, in amoeboid leukocytes, the MTOC is placed behind the nucleus toward the cell rear (Figure 2) (97, 98) and participates in the path finding mechanism. Once the cell’s nucleus, which represents the bulkiest part of the cell, and the associated MTOC successfully pass through a pore, protrusions directed to the smaller pores are retracted and the cell continues its movement through the path of least resistance. In this case, disrupting MTs leads to loss of cell coherency and results in fragmentation of the cell (97).

3 Microtubules and cell adhesive structures in invasion

One of the main distinctions between ameboid and mesenchymal migration is their adhesion-dependency. Unlike ameboid cells that do not require stable ECM attachment for their movement, mesenchymally migrating cells establish numerous interactions with the ECM, including the formation of integrin-based adhesions and proteolytically active structures that enable contact-driven invasion (Figure 2) (99101). Microtubules play a role in regulation and dynamics of these adhesive structures and are therefore an integral part of signaling regulating adhesion-dependent invasion.

3.1 Focal adhesions

FAs are integrin-based structures responsible for strong adhesion of cells to the ECM. Importantly, they also transmit information about the surrounding environment such as its stiffness by signaling to cytoskeleton associated proteins (102104). Both Rac and RhoA play role in FA regulation – whereas Rac activity is prominent in the formation of FAs, RhoA activity and Rac inhibition are needed for the maturation of FAs (105, 106).

The main role of MTs within FAs is the transport of integrins and metalloproteases (MMPs) to the cell membrane and the regulation of FAs turnover (107). Although there is no evidence of direct binding of MTs to FAs so far, MTs are known to be guided towards them and anchored in their proximity (108111). The targeting of mature FAs by MTs results in FA disassembly and cell edge retraction, and preventing the contact between MTs and FAs leads to enlarged FAs (112). In agreement, nocodazole-induced MTs depolymerization also results in larger FAs, whereas MT regrowth after nocodazole washout disassembles FAs (112114). Nevertheless, these studies were done in 2D environments, where the structure, composition and dynamics of FAs is different than in 3D environments (115), and many mechanisms valid in 2D systems are yet to be verified for 3D migration.

3.2 Podosomes

Podosomes and invadopodia are similar actin-based structures that play role in cell migration and ECM degradation, which is a key feature of mesenchymal invasion. Whereas podosomes are small, dot-like dynamic structures at the leading edge or organized rings exhibiting shallow ECM degradation, invadopodia are larger, irregularly shaped clusters usually localized in the central area of the cell. They are less dynamic and form outstretched extensions into the matrix resulting in deeper and more focused ECM degradation (116).

An intact MT network is necessary for the formation of podosomes since MT depolymerization (induced e.g. by nocodazole) leads to podosomal disassembly (117, 118). MTs are directed to podosomes through +TIPs such as EB1 and CLASPs (119, 120). Targeting of podosomes by MTs is associated with their higher dynamics, and podosomes without MTs show increased stability similarly to FAs (109, 112, 121).

The lifespan of MTs in podosomal structures is regulated by RhoGTPases. Inhibition of RhoA is able to increase the stability of MTs in podosomes, promoting podosome belt assembly (122). Accordingly, activation of RhoA leads to podosome disassembly through the RhoA/ROCK/MLCP axis as actomyosin contractility increases podosomes turnover and their disassembly (123125). Altering Cdc42 and Rac1 activity was also shown to disrupt podosomes (118, 122, 126).

3.3 Invadopodia

Invadopodia are invasive structures commonly found in various cancers (127). Unlike podosomes, microtubules are not required for invadopodia formation but are necessary for their elongation and correct function (128, 129). Invadopodia first form as a smaller actin-based structure with microtubules excluded from their core (130). Only after maturation of invadopodia, intermediate filaments and microtubules (typically 1-2 MTs per protrusion) invade their structure and allow invadopodia elongation. The microtubules that invade the shaft of invadopodia are stable, whereas at the base of the invadopodium more dynamic MTs are found (128). Accordingly, MT depolymerization does not affect the formation of small invadopodia, but limits their elongation and maturation (129).

One important role of MTs in invadopodia is the polarized trafficking of components such as matrix MMPs to the cell membrane (128, 131, 132). Moreover, the MT associated protein IQGAP1 accumulates in invadopodia where it interacts with exocyst components, and this interaction promotes invadopodia proteolysis by accumulation of MT1-MMP in a MT-regulated manner (133). Also, the exocytosis of MMP2 and MMP9 in melanoma cells is MTs-dependent (132) supporting the role of MTs in trafficking invadopodia components.

4 Microtubules and ECM conditions

It is well known that the conditions of the surrounding environment and its physical properties largely influence the choice of the migration mode. Amoeboid cells favor the large pores found in less dense ECM (134, 135) or confining conditions (100, 136), while mesenchymal cells with their proteolytic activity are able to migrate through stiff ECM and take advantage of the increased number of adhesion sites (101, 135, 137). Unsurprisingly, the MT network is receptive to ECM cues via posttranslational modifications that respond to ECM stiffness such as acetylation and glutamylation.

Generally, acetylated MTs are more stable and resilient. The acetylation of α tubulin is driven by α tubulin N-acetyltransferase 1 (αTAT1), and opposingly, histone deacetylase 6 (HDAC6) elicits tubulin deacetylation. Confusingly, both acetylation and deacetylation have been linked to accelerated cell migration. αTAT1 stabilizes MTs at the leading edge of migrating cells to promote cell motility and tumor progression (138). HDAC6 elicits pro-invasive signaling by activating Rho family GTPase, specifically by Rac1 (139). In addition to MTs, αTAT1 and HDAC6 are able to acetylate/deacetylate other substrates, such as cortactin. Cortactin contributes to actin filament assembly and is also required for MT1-MMP delivery to the leading edge of migrating cell (140), a process important for mesenchymal migration of tumor cells (141).

A recent study describes that ECM characteristics, MT dynamics and cell metabolism are interlinked to regulate cell invasion. Stiff substrates induce the conversion of glutamine to glutamate, increasing MT stability by glutamylation, resulting in an invasive phenotype and metastasis of breast cancer cells (142). However, a different study showed that higher density of collagen destabilizes MTs and induces GEFH1 mediated RhoA signaling (143).

Another characteristic of the surrounding environment that affects cell migration is oxygen availability. Hypoxia leads to MT depolymerization as a consequence of MAP4 and stathmin phosphorylation (144, 145). Subsequently, MTs depolymerization promotes RhoA activity by releasing GEFH1, a mechanism known to promote amoeboid invasion. In agreement, hypoxic conditions trigger the collective-amoeboid transition (146).

4.1 Microtubule targeting drugs

Microtubule drugs, due to their immense effect on MT structure and dynamics, have extensive impact on cell behavior. They are commonly used as chemotherapeutic agents based on their ability to arrest cell proliferation and cause cell death (147). Nevertheless, MT drugs exhibit more extensive behavior than just antimitotic effects, including deregulation of cell migration (148). They also interfere with invasion plasticity manifesting different effects on each invasion mode. Drugs inhibiting MT polymerization promote the amoeboid mode as it is less MT-dependent (Figure 2). For example, vincristine which is able to sequester tubulin dimers and prevent MT polymerization, induces the amoeboid phenotype through GEFH1/RhoA signaling (149). Similar results were observed with nocodazole, which also leads to MT disassembly and subsequent RhoA activation (150). In fibroblasts, nocodazole treatment prevented cells to adopt an elongated morphology and instead induced a round morphology, typical of the amoeboid phenotype (151). Treatment of cells with paclitaxel (taxol) stabilizes microtubules, but does not promote mesenchymal migration, instead it induces a non-motile phenotype (152154). In lymphocytes that utilize the amoeboid migration mode, taxol treatment inhibited migration in both 2D and microstructured environments, whereas nocodazole treatment increased membrane blebbing without increase in migration in 2D (155), but was able to promote amoeboid invasion in 3D (156).

The wide clinical usage of classic MT drugs is hampered by notable drug resistance and toxicity, fueling the chase for novel compounds with improved characteristics (157), many of which are in clinical trials (158). Moreover, in recent years it has become evident that the requirement for anti-metastatic behavior should be evaluated as well as primary growth shrinkage (159162). In point of fact, the prototypic antimitotic drugs paclitaxel and vincristine can in certain instances promote metastasis (163, 164). On the other hand, vinorelbine treatment in mice reduced metastasis more effectively than primary tumor growth (165). Another microtubule inhibitor, eribulin, exerts migrastatic behavior both in experimental conditions (166, 167) and in patients with advanced metastatic breast cancer (168).

Taken together, a suitable combination of microtubule drugs with invasion specific inhibitors could synergically target cell proliferation and both amoeboid and mesenchymal invasion, i.e., elicit both anti-proliferative and migrastatic effects.

5 Concluding remarks

The invasion phenotype is a result of the orchestration of all three cytoskeletal systems. Here, we have summarized evidence that MT dynamics can directly affect cancer invasion plasticity and described its role in both amoeboid and mesenchymal cells (Figure 2). Nevertheless, the contribution of the individual cytoskeletal components specifically during the transitions between amoeboid and mesenchymal invasion modes remains to be described. For example, it is not clear whether during MAT the disintegration of MTs precedes, follows, or accompanies formation of the actomyosin cortex. Moreover, the cells´ reaction to the rearrangement of the MTs network is dependent on several factors including differences between 2D and 3D environments, cell type, RhoGTPase signaling or presence of MT drugs, and thus the role of MTs in invasion is not uniform.

Above mentioned evidence shows that amoeboid migration is possible even if MTs are unstable or depleted and destabilization of MTs can directly induce amoeboid invasion. This is contrary to the finding that MTs are retained in amoeboid leukocytes and in fact promote migration by contributing to path-finding mechanisms or protrusion retraction (97, 169). These seemingly contradictory findings may be explained by the existence of multiple types of amoeboid invasion that include blebby, stable-bleb and pseudopodal subtypes that differ in their extent of adhesion, and protrusive and contractile activity, which occur based on cell type and/or ECM conditions (99, 170, 171). For example, leukocytes are known to adopt the pseudopodal amoeboid mode, which is dependent on MT and actin-driven pseudopod extension. On the other hand, the highly contractile bleb-based amoeboid modes are reliant on actomyosin activity, but do not require MTs (171) (Figure 2). Interestingly, the varying structure of the MT network in amoeboid cells is reflected also in Amoebozoa, unicellular protists after which the amoeboid migration mode is named. Based on immunocytochemistry staining of MTs, in some amoebae MTs are present as short cytoplasmic fibers, other contain long, parallel MT bundles and in some cases fibrous MTs where not detected at all (172). It thus seems that indeed forms of amoeboid migration dependent and independent on MT network exist.

On the other hand, mesenchymal migration requires the role of MT for multiple processes. Especially at the leading edge MTs contribute to extension and stabilization of protrusions, but also to formation of adhesive and proteolytic structures. However, pharmacological stabilization of MTs limits migration and invasion of cells, suggesting that excessive stabilization halts migration altogether.

In this context, it is not easy to conclude whether increased depolymerization of MTs leads to higher invasiveness or the opposite. On the contrary, what we can confirm is that MT dynamics directly affects the ability of the cell to choose among the invasive modes that are most profitable for them under the given conditions.

Author contributions

AŠ, DR, and JB contributed to the conception and processing of the article. AL, AŠ, and MP wrote the original manuscript. AL and AŠ contributed significantly to the first design of the images, AL created the images and all authors contributed to their finalization. All authors contributed to the article revision and approved the submitted version.

Funding

This work was supported by Charles University grant GAUK, project n. 922120 and by the National Institute for Cancer Research (Programme EXCELES, ID Project No. LX22NPO5102) - funded by the European Union - Next Generation EU. This work was also funded by Operational Programme Research, Development and Education, within the projects: Centre for Tumour Ecology—Research of the Cancer Microenvironment Supporting Cancer Growth and Spread (reg. No. CZ.02.1.01/0.0/0.0/16_019/0000785).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Alexandrova AY, Chikina AS, Svitkina TM. Actin cytoskeleton in mesenchymal-to-amoeboid transition of cancer cells. In: International review of cell and molecular biology. Elsevier (2020). p. 197–256. Available at: https://linkinghub.elsevier.com/retrieve/pii/S1937644820300782.

Google Scholar

2. Fife CM, McCarroll JA, Kavallaris M. Movers and shakers: cell cytoskeleton in cancer metastasis: Cytoskeleton and cancer metastasis. Br J Pharmacol (2014) 171(24):5507–23. doi: 10.1111/bph.12704

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Gandalovičová A, Vomastek T, Rosel D, Brábek J. Cell polarity signaling in the plasticity of cancer cell invasiveness. Oncotarget (2016) 7(18):25022–49. doi: 10.18632/oncotarget.7214

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Strouhalova K, Přechová M, Gandalovičová A, Brábek J, Gregor M, Rosel D. Vimentin intermediate filaments as potential target for cancer treatment. Cancers (2020) 12(1):184. doi: 10.3390/cancers12010184

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Paňková K, Rösel D, Novotný M, Brábek J. The molecular mechanisms of transition between mesenchymal and amoeboid invasiveness in tumor cells. Cell Mol Life Sci (2010) 67(1):63–71. doi: 10.1007/s00018-009-0132-1

PubMed Abstract | CrossRef Full Text | Google Scholar

6. te Boekhorst V, Friedl P. Plasticity of cancer cell invasion–mechanisms and implications for therapy. In: Advances in cancer research. Elsevier (2016). p. 209–64. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0065230X16300562.

Google Scholar

7. Tolde O, Gandalovičová A, Křížová A, Veselý P, Chmelík R, Rosel D, et al. Quantitative phase imaging unravels new insight into dynamics of mesenchymal and amoeboid cancer cell invasion. Sci Rep (2018) 8(1):12020. doi: 10.1038/s41598-018-30408-7

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Norman LL, Brugés J, Sengupta K, Sens P, Aranda-Espinoza H. Cell blebbing and membrane area homeostasis in spreading and retracting cells. Biophys J (2010) 99(6):1726–33. doi: 10.1016/j.bpj.2010.07.031

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Schick J, Raz E. Blebs–formation, regulation, positioning, and role in amoeboid cell migration. Front Cell Dev Biol (2022) 10:926394. doi: 10.3389/fcell.2022.926394

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Raftopoulou M, Hall A. Cell migration: Rho GTPases lead the way. Dev Biol (2004) 265(1):23–32. doi: 10.1016/j.ydbio.2003.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Eden S, Rohatgi R, Podtelejnikov AV, Mann M, Kirschner MW. Mechanism of regulation of WAVE1-induced actin nucleation by Rac1 and nck. Nature (2002) 418(6899):790–3. doi: 10.1038/nature00859

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Rohatgi R, Ho H yi H, Kirschner MW. Mechanism of n-wasp activation by Cdc42 and phosphatidylinositol 4,5-bisphosphate. J Cell Biol (2000) 150(6):1299–310. doi: 10.1083/jcb.150.6.1299

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Weaver AM, Young ME, Lee WL, Cooper JA. Integration of signals to the Arp2/3 complex. Curr Opin Cell Biol (2003) 15(1):23–30. doi: 10.1016/S0955-0674(02)00015-7

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Kraynov VS, Chamberlain C, Bokoch GM, Schwartz MA, Slabaugh S, Hahn KM. Localized rac activation dynamics visualized in living cells. Science (2000) 290(5490):333–7. doi: 10.1126/science.290.5490.333

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Alblas J, Ulfman L, Hordijk P, Koenderman L. Activation of RhoA and ROCK are essential for detachment of migrating Leukocytes Mol Biol Cell (2001) 12:9. doi: 10.1091/mbc.12.7.2137

CrossRef Full Text | Google Scholar

16. Amano M, Ito M, Kimura K, Fukata Y, Chihara K, Nakano T, et al. Phosphorylation and activation of myosin by rho-associated kinase (Rho-kinase). J Biol Chem (1996) 271(34):20246–9. doi: 10.1074/jbc.271.34.20246

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Kimura K, Ito M, Amano M, Chihara K, Fukata Y, Nakafuku M, et al. Regulation of myosin phosphatase by rho and rho-associated kinase (Rho-kinase). Science. (1996) 273(5272):245–8. doi: 10.1126/science.273.5272.245

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Maekawa M, Ishizaki T, Boku S, Watanabe N, Fujita A, Iwamatsu A, et al. Signaling from rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science (1999) 285(5429):895–8. doi: 10.1126/science.285.5429.895

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Kosla J, Paňková D, Plachý J, Tolde O, Bicanová K, Dvořák M, et al. Metastasis of aggressive amoeboid sarcoma cells is dependent on Rho/ROCK/MLC signaling. Cell Commun Signal (2013) 11(1):51. doi: 10.1186/1478-811X-11-51

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Orgaz JL, Herraiz C, Sanz-Moreno V. Rho GTPases modulate malignant transformation of tumor cells. Small GTPases (2014) 5(4):e983867. doi: 10.4161/sgtp.29019

CrossRef Full Text | Google Scholar

21. Čermák V, Gandalovičová A, Merta L, Harant K, Rösel D, Brábek J. High-throughput transcriptomic and proteomic profiling of mesenchymal-amoeboid transition in 3D collagen. Sci Data (2020) 7(1):160. doi: 10.1038/s41597-020-0499-2

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Sahai E, Marshall CJ. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol (2003) 5(8):711–9. doi: 10.1038/ncb1019

PubMed Abstract | CrossRef Full Text | Google Scholar

23. MacKay JL, Kumar S. Simultaneous and independent tuning of RhoA and Rac1 activity with orthogonally inducible promoters. Integr Biol (2014) 6(9):885–94. doi: 10.1039/c4ib00099d

CrossRef Full Text | Google Scholar

24. Sanz-Moreno V, Gadea G, Ahn J, Paterson H, Marra P, Pinner S, et al. Rac activation and inactivation control plasticity of tumor cell movement. Cell (2008) 135(3):510–23. doi: 10.1016/j.cell.2008.09.043

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Waterman-Storer CM, Worthylake RA, Liu BP, Burridge K, Salmon ED. Microtubule growth activates Rac1 to promote lamellipodial protrusion in fibroblasts. Nat Cell Biol (1999) 1(1):45–50. doi: 10.1038/9018

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Waterman-Storer CM, Salmon E. Positive feedback interactions between microtubule and actin dynamics during cell motility. Curr Opin Cell Biol (1999) 11(1):61–7. doi: 10.1016/S0955-0674(99)80008-8

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Daub H, Gevaert K, Vandekerckhove J, Sobel A, Hall A. Rac/Cdc42 and p65PAK regulate the microtubule-destabilizing protein stathmin through phosphorylation at serine 16. J Biol Chem (2001) 276(3):1677–80. doi: 10.1074/jbc.C000635200

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Zeitz M, Kierfeld J. Feedback mechanism for microtubule length regulation by stathmin gradients. Biophys J (2014) 107(12):2860–71. doi: 10.1016/j.bpj.2014.10.056

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Ishizaki T, Morishima Y, Okamoto M, Furuyashiki T, Kato T, Narumiya S. Coordination of microtubules and the actin cytoskeleton by the rho effector mDia1. Nat Cell Biol (2001) 3(1):8–14. doi: 10.1038/35050598

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Palazzo AF, Cook TA, Alberts AS, Gundersen GG. mDia mediates rho-regulated formation and orientation of stable microtubules. Nat Cell Biol (2001) 3(8):723–9. doi: 10.1038/35087035

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Sakamoto Y, Boëda B, Etienne-Manneville S. APC binds intermediate filaments and is required for their reorganization during cell migration. J Cell Biol (2013) 200(3):249–58. doi: 10.1083/jcb.201206010

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Schaedel L, Lorenz C, Schepers AV, Klumpp S, Köster S. Vimentin intermediate filaments stabilize dynamic microtubules by direct interactions. Nat Commun (2021) 12(1):3799. doi: 10.1038/s41467-021-23523-z

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Bouchet BP, Akhmanova A. Microtubules in 3D cell motility. J Cell Sci (2017) 130(1):39–50. doi: 10.1242/jcs.189431

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Etienne-Manneville S. Microtubules in cell migration. Annu Rev Cell Dev Biol (2013) 29(1):471–99. doi: 10.1146/annurev-cellbio-101011-155711

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Garcin C, Straube A. Microtubules in cell migration. Essays Biochem (2019) 63(5):509–20. doi: 10.1042/EBC20190016

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Vaughan KT. TIP maker and TIP marker; EB1 as a master controller of microtubule plus ends. J Cell Biol (2005) 171(2):197–200. doi: 10.1083/jcb.200509150

PubMed Abstract | CrossRef Full Text | Google Scholar

37. van Haren J, Charafeddine RA, Ettinger A, Wang H, Hahn KM, Wittmann T. Local control of intracellular microtubule dynamics by EB1 photodissociation. Nat Cell Biol (2018) 20(3):252–61. doi: 10.1038/s41556-017-0028-5

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Jayatilaka H, Giri A, Karl M, Aifuwa I, Trenton NJ, Phillip JM, et al. EB1 and cytoplasmic dynein mediate protrusion dynamics for efficient 3-dimensional cell migration. FASEB J (2018) 32(3):1207–21. doi: 10.1096/fj.201700444RR

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Bouchet BP, Noordstra I, van Amersfoort M, Katrukha EA, Ammon YC, ter Hoeve ND, et al. Mesenchymal cell invasion requires cooperative regulation of persistent microtubule growth by SLAIN2 and CLASP1. Dev Cell (2016) 39(6):708–23. doi: 10.1016/j.devcel.2016.11.009

PubMed Abstract | CrossRef Full Text | Google Scholar

40. White CD, Erdemir HH, Sacks DB. IQGAP1 and its binding proteins control diverse biological functions. Cell Signal (2012) 24(4):826–34. doi: 10.1016/j.cellsig.2011.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Bashour AM, Fullerton AT, Hart MJ, Bloom GS. IQGAP1, a rac- and Cdc42-binding protein, directly binds and cross-links microfilaments. J Cell Biol (1997) 137(7):1555–66. doi: 10.1083/jcb.137.7.1555

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Hoeprich GJ, Sinclair AN, Shekhar S, Goode BL. Single-molecule imaging of IQGAP1 regulating actin filament dynamics. Mol Biol Cell (2022) 33(1):ar2. doi: 10.1091/mbc.E21-04-0211

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Kuroda S, Fukata M, Kobayashi K, Nakafuku M, Nomura N, Iwamatsu A, et al. Identification of IQGAP as a putative target for the small GTPases, Cdc42 and Rac1. J Biol Chem (1996) 271(38):23363–7. doi: 10.1074/jbc.271.38.23363

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Fukata M, Kuroda S, Fujii K, Nakamura T, Shoji I, Matsuura Y, et al. Regulation of cross-linking of actin filament by IQGAP1, a target for Cdc42. J Biol Chem (1997) 272(47):29579–83. doi: 10.1074/jbc.272.47.29579

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Benseñor LB, Kan HM, Wang N, Wallrabe H, Davidson LA, Cai Y, et al. IQGAP1 regulates cell motility by linking growth factor signaling to actin assembly. J Cell Sci (2007) 120(4):658–69. doi: 10.1242/jcs.03376

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Le Clainche C, Schlaepfer D, Ferrari A, Klingauf M, Grohmanova K, Veligodskiy A, et al. IQGAP1 stimulates actin assembly through the n-Wasp-Arp2/3 pathway. J Biol Chem (2007) 282(1):426–35. doi: 10.1074/jbc.M607711200

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Mataraza JM, Briggs MW, Li Z, Entwistle A, Ridley AJ, Sacks DB. IQGAP1 promotes cell motility and invasion. J Biol Chem (2003) 278(42):41237–45. doi: 10.1074/jbc.M304838200

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Hart MJ, Callow MG, Souza B, Polakis P. IQGAP1, a calmodulin-binding protein with a rasGAP-related domain, is a potential effector for cdc42Hs. EMBO J (1996) 15(12):2997–3005. doi: 10.1002/j.1460-2075.1996.tb00663.x

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Etienne-Manneville S, Hall A. Integrin-mediated activation of Cdc42 controls cell polarity in migrating astrocytes through PKCζ. Cell (2001) 106(4):489–98. doi: 10.1016/S0092-8674(01)00471-8

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Fukata M, Watanabe T, Noritake J, Nakagawa M, Yamaga M, Kuroda S, et al. Rac1 and Cdc42 capture microtubules through IQGAP1 and CLIP-170. Cell (2002) 109(7):873–85. doi: 10.1016/S0092-8674(02)00800-0

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Watanabe T, Wang S, Noritake J, Sato K, Fukata M, Takefuji M, et al. Interaction with IQGAP1 links APC to Rac1, Cdc42, and actin filaments during cell polarization and migration. Dev Cell (2004) 7(6):871–83. doi: 10.1016/j.devcel.2004.10.017

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Cao D, Su Z, Wang W, Wu H, Liu X, Akram S, et al. Signaling scaffold protein IQGAP1 interacts with microtubule plus-end tracking protein SKAP and links dynamic microtubule plus-end to steer cell migration. J Biol Chem (2015) 290(39):23766–80. doi: 10.1074/jbc.M115.673517

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Yamaoka-Tojo M, Ushio-Fukai M, Hilenski L, Dikalov SI, Chen YE, Tojo T, et al. IQGAP1, a novel vascular endothelial growth factor receptor binding protein, is involved in reactive oxygen species–dependent endothelial migration and proliferation. Circ Res (2004) 95(3):276–83. doi: 10.1161/01.RES.0000136522.58649.60

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Li S, Guan JL, Chien S. Biochemistry and biomechanics of cell motility. Annu Rev BioMed Eng (2005) 7(1):105–50. doi: 10.1146/annurev.bioeng.7.060804.100340

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Sánchez-Huertas C, Bonhomme M, Falco A, Fagotto-Kaufmann C, van Haren J, Jeanneteau F, et al. The +TIP navigator-1 is an actin–microtubule crosslinker that regulates axonal growth cone motility. J Cell Biol (2020) 219(9):e201905199. doi: 10.1083/jcb.201905199

PubMed Abstract | CrossRef Full Text | Google Scholar

56. van Haren J, Draegestein K, Keijzer N, Abrahams JP, Grosveld F, Peeters PJ, et al. Mammalian navigators are microtubule plus-end tracking proteins that can reorganize the cytoskeleton to induce neurite-like extensions. Cell Motil Cytoskeleton (2009) 66(10):824–38. doi: 10.1002/cm.20370

PubMed Abstract | CrossRef Full Text | Google Scholar

57. van Haren J, Boudeau J, Schmidt S, Basu S, Liu Z, Lammers D, et al. Dynamic microtubules catalyze formation of navigator-TRIO complexes to regulate neurite extension. Curr Biol (2014) 24(15):1778–85. doi: 10.1016/j.cub.2014.06.037

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Bellanger JM, Lazaro JB, Diriong S, Fernandez A, Lamb N, Debant A. The two guanine nucleotide exchange factor domains of trio link the Rac1 and the RhoA pathways in vivo. Oncogene (1998) 16(2):147–52. doi: 10.1038/sj.onc.1201532

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Briançon-Marjollet A, Ghogha A, Nawabi H, Triki I, Auziol C, Fromont S, et al. Trio mediates netrin-1-Induced Rac1 activation in axon outgrowth and guidance. Mol Cell Biol (2008) 28(7):2314–23. doi: 10.1128/MCB.00998-07

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Cohen-Dvashi H, Ben-Chetrit N, Russell R, Carvalho S, Lauriola M, Nisani S, et al. Navigator-3, a modulator of cell migration, may act as a suppressor of breast cancer progression. EMBO Mol Med (2015) 7(3):299–314. doi: 10.15252/emmm.201404134

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Belmont LD, Mitchison TJ. Identification of a protein that interacts with tubulin dimers and increases the catastrophe rate of microtubules. Cell (1996) 84(4):623–31. doi: 10.1016/S0092-8674(00)81037-5

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Curmi PA, Andersen SSL, Lachkar S, Gavet O, Karsenti E, Knossow M, et al. The Stathmin/Tubulin interaction in vitro. J Biol Chem (1997) 272(40):25029–36. doi: 10.1074/jbc.272.40.25029

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Howell B, Larsson N, Gullberg M, Cassimeris L. Dissociation of the tubulin-sequestering and microtubule catastrophe-promoting activities of oncoprotein 18/Stathmin. Mol Biol Cell (1999) 10(1):105–18. doi: 10.1091/mbc.10.1.105

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Melander Gradin H, Marklund U, Larsson N, Chatila TA, Gullberg M. Regulation of microtubule dynamics by Ca2+/calmodulin-dependent kinase IV/Gr-dependent phosphorylation of oncoprotein 18. Mol Cell Biol (1997) 17(6):3459–67. doi: 10.1128/MCB.17.6.3459

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Gavet O, Ozon S, Manceau V, Lawler S, Curmi P, Sobel A. The stathmin phosphoprotein family: intracellular localization and effects on the microtubule network. J Cell Sci (1998) 111(22):3333–46. doi: 10.1242/jcs.111.22.3333

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Ng DCH, Lin BH, Lim CP, Huang G, Zhang T, Poli V, et al. Stat3 regulates microtubules by antagonizing the depolymerization activity of stathmin. J Cell Biol (2006) 172(2):245–57. doi: 10.1083/jcb.200503021

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Baldassarre G, Belletti B, Nicoloso MS, Schiappacassi M, Vecchione A, Spessotto P, et al. p27Kip1-stathmin interaction influences sarcoma cell migration and invasion. Cancer Cell (2005) 7(1):51–63. doi: 10.1016/j.ccr.2004.11.025

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Schiappacassi M, Lovat F, Canzonieri V, Belletti B, Berton S, Di Stefano D, et al. p27 Kip1 expression inhibits glioblastoma growth, invasion, and tumor-induced neoangiogenesis. Mol Cancer Ther (2008) 7(5):1164–75. doi: 10.1158/1535-7163.MCT-07-2154

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Klausen P, Pedersen L, Jurlander J, Baumann H. Oncostatin m and interleukin 6 inhibit cell cycle progression by prevention of p27kip1 degradation in HepG2 cells. Oncogene (2000) 19(32):3675–83. doi: 10.1038/sj.onc.1203707

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Niethammer P, Bastiaens P, Karsenti E. Stathmin-tubulin interaction gradients in motile and mitotic cells. Science (2004) 303(5665):1862–6. doi: 10.1126/science.1094108

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Belletti B, Nicoloso MS, Schiappacassi M, Berton S, Lovat F, Wolf K, et al. Stathmin activity influences sarcoma cell shape, motility, and metastatic potential. Mol Biol Cell (2008) 19(5):2003–13. doi: 10.1091/mbc.e07-09-0894

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Baldassarre G, Belletti B, Bruni P, Boccia A, Trapasso F, Pentimalli F, et al. Overexpressed cyclin D3 contributes to retaining the growth inhibitor p27 in the cytoplasm of thyroid tumor cells. J Clin Invest (1999) 104(7):865–74. doi: 10.1172/JCI6443

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Besson A, Gurian-West M, Schmidt A, Hall A, Roberts JM. p27 Kip1 modulates cell migration through the regulation of RhoA activation. Genes Dev (2004) 18(8):862–76. doi: 10.1101/gad.1185504

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Morelli G, Even A, Gladwyn-Ng I, Le Bail R, Shilian M, Godin JD, et al. p27Kip1 modulates axonal transport by regulating α-tubulin acetyltransferase 1 stability. Cell Rep (2018) 23(8):2429–42. doi: 10.1016/j.celrep.2018.04.083

PubMed Abstract | CrossRef Full Text | Google Scholar

75. McAllister SS, Becker-Hapak M, Pintucci G, Pagano M, Dowdy SF. Novel p27 kip1 c-terminal scatter domain mediates rac-dependent cell migration independent of cell cycle arrest functions. Mol Cell Biol (2003) 23(1):216–28. doi: 10.1128/MCB.23.1.216-228.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Berton S, Belletti B, Wolf K, Canzonieri V, Lovat F, Vecchione A, et al. The tumor suppressor functions of p27 kip1 include control of the Mesenchymal/Amoeboid transition. Mol Cell Biol (2009) 29(18):5031–45. doi: 10.1128/MCB.00144-09

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Gui P, Labrousse A, Van Goethem E, Besson A, Maridonneau-Parini I, Le Cabec V. Rho/ROCK pathway inhibition by CDK inhibitor p27kip1 participates in the onset of macrophage 3D-mesenchymal migration. J Cell Sci (2014) 127(18):4009–23. doi: 10.1242/jcs.150987

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Chalkia D, Nikolaidis N, Makalowski W, Klein J, Nei M. Origins and evolution of the formin multigene family that is involved in the formation of actin filaments. Mol Biol Evol (2008) 25(12):2717–33. doi: 10.1093/molbev/msn215

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Hager MH, Morley S, Bielenberg DR, Gao S, Morello M, Holcomb IN, et al. DIAPH3 governs the cellular transition to the amoeboid tumour phenotype. EMBO Mol Med (2012) 4(8):743–60. doi: 10.1002/emmm.201200242

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Wyse MM, Goicoechea S, Garcia-Mata R, Nestor-Kalinoski AL, Eisenmann KM. mDia2 and CXCL12/CXCR4 chemokine signaling intersect to drive tumor cell amoeboid morphological transitions. Biochem Biophys Res Commun (2017) 484(2):255–61. doi: 10.1016/j.bbrc.2017.01.087

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Pettee KM, Dvorak KM, Nestor-Kalinoski AL, Eisenmann KM. An mDia2/ROCK signaling axis regulates invasive egress from epithelial ovarian cancer spheroids. Aspenstrom P editor PloS One (2014) 9(2):e90371. doi: 10.1371/journal.pone.0090371

CrossRef Full Text | Google Scholar

82. Morley S, You S, Pollan S, Choi J, Zhou B, Hager MH, et al. Regulation of microtubule dynamics by DIAPH3 influences amoeboid tumor cell mechanics and sensitivity to taxanes. Sci Rep (2015) 5(1):12136. doi: 10.1038/srep12136

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Nalbant P, Chang YC, Birkenfeld J, Chang ZF, Bokoch GM. Guanine nucleotide exchange factor-H1 regulates cell migration via localized activation of RhoA at the leading edge. Forscher P editor Mol Biol Cell (2009) 20(18):4070–82. doi: 10.1091/mbc.e09-01-0041

CrossRef Full Text | Google Scholar

84. Azoitei ML, Noh J, Marston DJ, Roudot P, Marshall CB, Daugird TA, et al. Spatiotemporal dynamics of GEF-H1 activation controlled by microtubule- and src-mediated pathways. J Cell Biol (2019) 218(9):3077–97. doi: 10.1083/jcb.201812073

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Pan YR, Chen CC, Chan YT, Wang HJ, Chien FT, Chen YL, et al. STAT3-coordinated migration facilitates the dissemination of diffuse large b-cell lymphomas. Nat Commun (2018) 9(1):3696. doi: 10.1038/s41467-018-06134-z

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Soltani MH, Pichardo R, Song Z, Sangha N, Camacho F, Satyamoorthy K, et al. Microtubule-associated protein 2, a marker of neuronal differentiation, induces mitotic defects, inhibits growth of melanoma cells, and predicts metastatic potential of cutaneous melanoma. Am J Pathol (2005) 166(6):1841–50. doi: 10.1016/S0002-9440(10)62493-5

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Ou Y, Zheng X, Gao Y, Shu M, Leng T, Li Y, et al. Activation of cyclic AMP/PKA pathway inhibits bladder cancer cell invasion by targeting MAP4-dependent microtubule dynamics. Urol Oncol Semin Orig Investig (2014) 32(1):47.e21–8. doi: 10.1016/j.urolonc.2013.06.017

CrossRef Full Text | Google Scholar

88. Jiang YY, Shang L, Shi ZZ, Zhang TT, Ma S, Lu CC, et al. Microtubule-associated protein 4 is an important regulator of cell invasion/migration and a potential therapeutic target in esophageal squamous cell carcinoma. Oncogene (2016) 35(37):4846–56. doi: 10.1038/onc.2016.17

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Tortosa E, Montenegro-Venegas C, Benoist M, Härtel S, González-Billault C, Esteban JA, et al. Microtubule-associated protein 1B (MAP1B) is required for dendritic spine development and synaptic maturation. J Biol Chem (2011) 286(47):40638–48. doi: 10.1074/jbc.M111.271320

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Chang W, Gruber D, Chari S, Kitazawa H, Hamazumi Y, Hisanaga S, et al. Phosphorylation of MAP4 affects microtubule properties and cell cycle progression. J Cell Sci (2001) 114(15):2879–87. doi: 10.1242/jcs.114.15.2879

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Krendel M, Zenke FT, Bokoch GM. Nucleotide exchange factor GEF-H1 mediates cross-talk between microtubules and the actin cytoskeleton. Nat Cell Biol (2002) 4(4):294–301. doi: 10.1038/ncb773

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Luxton GG, Gundersen GG. Orientation and function of the nuclear–centrosomal axis during cell migration. Curr Opin Cell Biol (2011) 23(5):579–88. doi: 10.1016/j.ceb.2011.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Burute M, Prioux M, Blin G, Truchet S, Letort G, Tseng Q, et al. Polarity reversal by centrosome repositioning primes cell scattering during epithelial-to-Mesenchymal transition. Dev Cell (2017) 40(2):168–84. doi: 10.1016/j.devcel.2016.12.004

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Doyle AD, Wang FW, Matsumoto K, Yamada KM. One-dimensional topography underlies three-dimensional fibrillar cell migration. J Cell Biol (2009) 184(4):481–90. doi: 10.1083/jcb.200810041

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Zhang J, Wang Y. Centrosome defines the rear of cells during mesenchymal migration. Mol Biol Cell (2017) 28(23):3240–51. doi: 10.1091/mbc.e17-06-0366

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Pouthas F, Girard P, Lecaudey V, Ly TBN, Gilmour D, Boulin C, et al. In migrating cells, the golgi complex and the position of the centrosome depend on geometrical constraints of the substratum. J Cell Sci (2008) 121(14):2406–14. doi: 10.1242/jcs.026849

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Renkawitz J, Kopf A, Stopp J, de Vries I, Driscoll MK, Merrin J, et al. Nuclear positioning facilitates amoeboid migration along the path of least resistance. Nature. (2019) 568(7753):546–50. doi: 10.1038/s41586-019-1087-5

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Sánchez-Madrid F, Serrador JM. Bringing up the rear: defining the roles of the uropod. Nat Rev Mol Cell Biol (2009) 10(5):353–9. doi: 10.1038/nrm2680

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Friedl P, Wolf K. Plasticity of cell migration: A multiscale tuning model. J Cell Biol (2010) 188(1):11–9. doi: 10.1083/jcb.200909003

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Liu YJ, Le Berre M, Lautenschlaeger F, Maiuri P, Callan-Jones A, Heuzé M, et al. Confinement and low adhesion induce fast amoeboid migration of slow mesenchymal cells. Cell (2015) 160(4):659–72. doi: 10.1016/j.cell.2015.01.007

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Wolf K, Mazo I, Leung H, Engelke K, von Andrian UH, Deryugina EI, et al. Compensation mechanism in tumor cell migration. J Cell Biol (2003) 160(2):267–77. doi: 10.1083/jcb.200209006

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Doyle AD, Carvajal N, Jin A, Matsumoto K, Yamada KM. Local 3D matrix microenvironment regulates cell migration through spatiotemporal dynamics of contractility-dependent adhesions. Nat Commun (2015) 6(1):8720. doi: 10.1038/ncomms9720

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Seetharaman S, Etienne-Manneville S. Integrin diversity brings specificity in mechanotransduction: Integrin diversity brings specificity in mechanotransduction. Biol Cell (2018) 110(3):49–64. doi: 10.1111/boc.201700060

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Wozniak MA, Modzelewska K, Kwong L, Keely PJ. Focal adhesion regulation of cell behavior. Biochim Biophys Acta BBA - Mol Cell Res (2004) 1692(2–3):103–19. doi: 10.1016/j.bbamcr.2004.04.007

CrossRef Full Text | Google Scholar

105. Huveneers S, Danen EHJ. Adhesion signaling – crosstalk between integrins, src and rho. J Cell Sci (2009) 122(8):1059–69. doi: 10.1242/jcs.039446

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Lawson CD, Burridge K. The on-off relationship of rho and rac during integrin-mediated adhesion and cell migration. Small GTPases (2014) 5(1):e27958. doi: 10.4161/sgtp.27958

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Stehbens S, Wittmann T. Targeting and transport: How microtubules control focal adhesion dynamics. J Cell Biol (2012) 198(4):481–9. doi: 10.1083/jcb.201206050

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Kaverina I, Rottner K, Small JV. Targeting, capture, and stabilization of microtubules at early focal adhesions. J Cell Biol (1998) 142(1):181–90. doi: 10.1083/jcb.142.1.181

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Krylyshkina O, Anderson KI, Kaverina I, Upmann I, Manstein DJ, Small JV, et al. Nanometer targeting of microtubules to focal adhesions. J Cell Biol (2003) 161(5):853–9. doi: 10.1083/jcb.200301102

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Seetharaman S, Etienne-Manneville S. Microtubules at focal adhesions – a double-edged sword. J Cell Sci (2019) 132(19):jcs232843. doi: 10.1242/jcs.232843

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Byron A, Askari JA, Humphries JD, Jacquemet G, Koper EJ, Warwood S, et al. A proteomic approach reveals integrin activation state-dependent control of microtubule cortical targeting. Nat Commun (2015) 6(1):6135. doi: 10.1038/ncomms7135

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Kaverina I, Krylyshkina O, Small JV. Microtubule targeting of substrate contacts promotes their relaxation and dissociation. J Cell Biol (1999) 146(5):1033–44. doi: 10.1083/jcb.146.5.1033

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Bershadsky A, Chausovsky A, Becker E, Lyubimova A, Geiger B. Involvement of microtubules in the control of adhesion-dependent signal transduction. Curr Biol (1996) 6(10):1279–89. doi: 10.1016/S0960-9822(02)70714-8

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Ezratty EJ, Partridge MA, Gundersen GG. Microtubule-induced focal adhesion disassembly is mediated by dynamin and focal adhesion kinase. Nat Cell Biol (2005) 7(6):581–90. doi: 10.1038/ncb1262

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Tolde O, Rösel D, Janostiak R, Vesely P, Brábek J. Dynamics and morphology of focal adhesions in complex 3D environment. Folia Biol (Praha) (2012) 58:177–84.

PubMed Abstract | Google Scholar

116. Linder S. The matrix corroded: podosomes and invadopodia in extracellular matrix degradation. Trends Cell Biol (2007) 17(3):107–17. doi: 10.1016/j.tcb.2007.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Linder S, Hufner K, Wintergerst U, Aepfelbacher M. Microtubule-dependent formation of podosomal adhesion structures in primary human macrophages. J Cell Sci (2000) 113(23):4165–76. doi: 10.1242/jcs.113.23.4165

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Ory S, Destaing O, Jurdic P. Microtubule dynamics differentially regulates rho and rac activity and triggers rho-independent stress fiber formation in macrophage polykaryons. Eur J Cell Biol (2002) 81(6):351–62. doi: 10.1078/0171-9335-00255

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Biosse Duplan M, Zalli D, Stephens S, Zenger S, Neff L, Oelkers JM, et al. Microtubule dynamic instability controls podosome patterning in osteoclasts through EB1, cortactin, and src. Mol Cell Biol (2014) 34(1):16–29. doi: 10.1128/MCB.00578-13

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Efimova N, Grimaldi A, Bachmann A, Frye K, Zhu X, Feoktistov A, et al. Podosome-regulating kinesin KIF1C translocates to the cell periphery in a CLASP-dependent manner. J Cell Sci (2014) 127(24):5179–88. doi: 10.1242/jcs.149633

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Kopp P, Lammers R, Aepfelbacher M, Rudel T, Machuy N, Steffen W, et al. The kinesin KIF1C and microtubule plus ends regulate podosome dynamics in Macrophages□D □V. Mol Biol Cell (2006) 17:13. doi: 10.1091/mbc.e05-11-1010

CrossRef Full Text | Google Scholar

122. Destaing O, Saltel F, Gilquin B, Chabadel A, Khochbin S, Ory S, et al. A novel rho-mDia2-HDAC6 pathway controls podosome patterning through microtubule acetylation in osteoclasts. J Cell Sci (2005) 118(13):2901–11. doi: 10.1242/jcs.02425

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Bhuwania R, Cornfine S, Fang Z, Krüger M, Luna EJ, Linder S. Supervillin couples myosin-dependent contractility to podosomes and enables their turnover. J Cell Sci (2012) 125(9):2300–14. doi: 10.1242/jcs.100032

PubMed Abstract | CrossRef Full Text | Google Scholar

124. van den Dries K, Meddens MBM, de Keijzer S, Shekhar S, Subramaniam V, Figdor CG, et al. Interplay between myosin IIA-mediated contractility and actin network integrity orchestrates podosome composition and oscillations. Nat Commun (2013) 4(1):1412. doi: 10.1038/ncomms2402

PubMed Abstract | CrossRef Full Text | Google Scholar

125. van Helden SFG, Oud MM, Joosten B, Peterse N, Figdor CG, van Leeuwen FN. PGE2-mediated podosome loss in dendritic cells is dependent on actomyosin contraction downstream of the RhoA–rho-kinase axis. J Cell Sci (2008) 121(7):1096–106. doi: 10.1242/jcs.020289

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Ory S, Brazier H, Pawlak G, Blangy A. Rho GTPases in osteoclasts: Orchestrators of podosome arrangement. Eur J Cell Biol (2008) 87(8–9):469–77. doi: 10.1016/j.ejcb.2008.03.002

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Gimona M, Buccione R, Courtneidge SA, Linder S. Assembly and biological role of podosomes and invadopodia. Curr Opin Cell Biol (2008) 20(2):235–41. doi: 10.1016/j.ceb.2008.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Schoumacher M, Goldman RD, Louvard D, Vignjevic DM. Actin, microtubules, and vimentin intermediate filaments cooperate for elongation of invadopodia. J Cell Biol (2010) 189(3):541–56. doi: 10.1083/jcb.200909113

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Kikuchi K, Takahashi K. WAVE2- and microtubule-dependent formation of long protrusions and invasion of cancer cells cultured on three-dimensional extracellular matrices. Cancer Sci (2008) 99(11):2252–9. doi: 10.1111/j.1349-7006.2008.00927.x

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Revach OY, Weiner A, Rechav K, Sabanay I, Livne A, Geiger B. Mechanical interplay between invadopodia and the nucleus in cultured cancer cells. Sci Rep (2015) 5(1):9466. doi: 10.1038/srep09466

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Caldieri G, Buccione R. Aiming for invadopodia: organizing polarized delivery at sites of invasion. Trends Cell Biol (2010) 20(2):64–70. doi: 10.1016/j.tcb.2009.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Schnaeker EM, Ossig R, Ludwig T, Dreier R, Oberleithner H, Wilhelmi M, et al. Microtubule-dependent matrix metalloproteinase-2/Matrix metalloproteinase-9 exocytosis. Cancer Res (2004) 64(24):8924–31. doi: 10.1158/0008-5472.CAN-04-0324

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Sakurai-Yageta M, Recchi C, Le Dez G, Sibarita JB, Daviet L, Camonis J, et al. The interaction of IQGAP1 with the exocyst complex is required for tumor cell invasion downstream of Cdc42 and RhoA. J Cell Biol (2008) 181(6):985–98. doi: 10.1083/jcb.200709076

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Brábek J, Mierke CT, Rösel D, Veselý P, Fabry B. The role of the tissue microenvironment in the regulation of cancer cell motility and invasion. Cell Commun Signal (2010) 8(1):22. doi: 10.1186/1478-811X-8-22

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Wolf K, te Lindert M, Krause M, Alexander S, te Riet J, Willis AL, et al. Physical limits of cell migration: Control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J Cell Biol (2013) 201(7):1069–84. doi: 10.1083/jcb.201210152

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Ruprecht V, Wieser S, Callan-Jones A, Smutny M, Morita H, Sako K, et al. Cortical contractility triggers a stochastic switch to fast amoeboid cell motility. Cell (2015) 160(4):673–85. doi: 10.1016/j.cell.2015.01.008

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Plotnikov SV, Waterman CM. Guiding cell migration by tugging. Curr Opin Cell Biol (2013) 25(5):619–26. doi: 10.1016/j.ceb.2013.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Montagnac G, Meas-Yedid V, Irondelle M, Castro-Castro A, Franco M, Shida T, et al. αTAT1 catalyses microtubule acetylation at clathrin-coated pits. Nature (2013) 502(7472):567–70. doi: 10.1038/nature12571

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Pham TQ, Robinson K, Xu L, Pavlova MN, Skapek SX, Chen EY. HDAC6 promotes growth, migration/invasion, and self-renewal of rhabdomyosarcoma. Oncogene (2021) 40(3):578–91. doi: 10.1038/s41388-020-01550-2

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Clark ES, Whigham AS, Yarbrough WG, Weaver AM. Cortactin is an essential regulator of matrix metalloproteinase secretion and extracellular matrix degradation in invadopodia. Cancer Res (2007) 67(9):4227–35. doi: 10.1158/0008-5472.CAN-06-3928

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Castro-Castro A, Janke C, Montagnac G, Paul-Gilloteaux P, Chavrier P. ATAT1/MEC-17 acetyltransferase and HDAC6 deacetylase control a balance of acetylation of alpha-tubulin and cortactin and regulate MT1-MMP trafficking and breast tumor cell invasion. Eur J Cell Biol (2012) 91(11–12):950–60. doi: 10.1016/j.ejcb.2012.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Torrino S, Grasset EM, Audebert S, Belhadj I, Lacoux C, Haynes M, et al. Mechano-induced cell metabolism promotes microtubule glutamylation to force metastasis. Cell Metab (2021) 33(7):1342–57. doi: 10.1016/j.cmet.2021.05.009

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Heck JN, Ponik SM, Garcia-Mendoza MG, Pehlke CA, Inman DR, Eliceiri KW, et al. Microtubules regulate GEF-H1 in response to extracellular matrix stiffness. Mol Biol Cell (2012) 23(13):2583–92. doi: 10.1091/mbc.e11-10-0876

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Hu JY, Chu ZG, Han J, Dang Y, Yan H, Zhang Q, et al. The p38/MAPK pathway regulates microtubule polymerization through phosphorylation of MAP4 and Op18 in hypoxic cells. Cell Mol Life Sci (2010) 67(2):321–33. doi: 10.1007/s00018-009-0187-z

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Zhang J, Li L, Zhang Q, Yang X, Zhang C, Zhang X, et al. Phosphorylation of microtubule- associated protein 4 promotes hypoxic endothelial cell migration and proliferation. Front Pharmacol (2019) 10:368. doi: 10.3389/fphar.2019.00368

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Lehmann S, te Boekhorst V, Odenthal J, Bianchi R, van Helvert S, Ikenberg K, et al. Hypoxia induces a HIF-1-Dependent transition from collective-to-Amoeboid dissemination in epithelial cancer cells. Curr Biol (2017) 27(3):392–400. doi: 10.1016/j.cub.2016.11.057

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Čermák V, Dostál V, Jelínek M, Libusová L, Kovář J, Rösel D, et al. Microtubule-targeting agents and their impact on cancer treatment. Eur J Cell Biol (2020) 99(4):151075. doi: 10.1016/j.ejcb.2020.151075

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Bates D, Eastman A. Microtubule destabilising agents: far more than just antimitotic anticancer drugs: MDA mechanisms of action. Br J Clin Pharmacol (2017) 83(2):255–68. doi: 10.1111/bcp.13126

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Eitaki M, Yamamori T, Meike S, Yasui H, Inanami O. Vincristine enhances amoeboid-like motility via GEF-H1/RhoA/ROCK/Myosin light chain signaling in MKN45 cells. BMC Cancer (2012) 12(1):469. doi: 10.1186/1471-2407-12-469

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Chang YC, Nalbant P, Birkenfeld J, Chang ZF, Bokoch GM. GEF-H1 couples nocodazole-induced microtubule disassembly to cell contractility via RhoA. Mol Biol Cell (2008) 19(5):2147–53. doi: 10.1091/mbc.e07-12-1269

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Belletti B, Pellizzari I, Berton S, Fabris L, Wolf K, Lovat F, et al. p27 kip1 controls cell morphology and motility by regulating microtubule-dependent lipid raft recycling. Mol Cell Biol (2010) 30(9):2229–40. doi: 10.1128/MCB.00723-09

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Carey SP, Rahman A, Kraning-Rush CM, Romero B, Somasegar S, Torre OM, et al. Comparative mechanisms of cancer cell migration through 3D matrix and physiological microtracks. Am J Physiol-Cell Physiol (2015) 308(6):C436–47. doi: 10.1152/ajpcell.00225.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

153. Tran TA, Gillet L, Roger S, Besson P, White E, Le Guennec JY. Non-anti-mitotic concentrations of taxol reduce breast cancer cell invasiveness. Biochem Biophys Res Commun (2009) 379(2):304–8. doi: 10.1016/j.bbrc.2008.12.073

PubMed Abstract | CrossRef Full Text | Google Scholar

154. Yvon AMC, Wadsworth P, Jordan MA. Taxol suppresses dynamics of individual microtubules in living human tumor cells. Mol Biol Cell (1999) 10(4):947–59. doi: 10.1091/mbc.10.4.947

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Takesono A, Heasman SJ, Wojciak-Stothard B, Garg R, Ridley AJ. Microtubules regulate migratory polarity through Rho/ROCK signaling in T cells. PloS One (2010) 5(1):e8774. doi: 10.1371/journal.pone.0008774

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Tabdanov ED, Rodríguez-Merced NJ, Cartagena-Rivera AX, Puram VV, Callaway MK, Ensminger EA, et al. Engineering T cells to enhance 3D migration through structurally and mechanically complex tumor microenvironments. Nat Commun (2021) 12(1):2815. doi: 10.1038/s41467-021-22985-5

PubMed Abstract | CrossRef Full Text | Google Scholar

157. Dumontet C, Jordan MA. Microtubule-binding agents: A dynamic field of cancer therapeutics. Nat Rev Drug Discov (2010) 9(10):790–803. doi: 10.1038/nrd3253

PubMed Abstract | CrossRef Full Text | Google Scholar

158. Škubník J, Jurášek M, Ruml T, Rimpelová S. Mitotic poisons in research and medicine. Molecules (2020) 25(20):4632. doi: 10.3390/molecules25204632

PubMed Abstract | CrossRef Full Text | Google Scholar

159. Fernandes M, Rosel D, Brábek J. Translation in solid cancer: are size-based response criteria an anachronism? Clin Transl Oncol (2015) 17(1):1–10. doi: 10.1007/s12094-014-1207-5

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Gandalovičová A, Rosel D, Fernandes M, Veselý P, Heneberg P, Čermák V, et al. Migrastatics–anti-metastatic and anti-invasion drugs: Promises and challenges. Trends Cancer (2017) 3(6):391–406. doi: 10.1016/j.trecan.2017.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

161. Rosel D, Fernandes M, Sanz-Moreno V, Brábek J. Migrastatics: Redirecting R&D in solid cancer towards metastasis? Trends Cancer (2019) 5(12):755–6. doi: 10.1016/j.trecan.2019.10.011

PubMed Abstract | CrossRef Full Text | Google Scholar

162. Solomon J, Raškova M, Rösel D, Brábek J, Gil-Henn H. Are we ready for migrastatics? Cells (2021) 10(8):1845. doi: 10.3390/cells10081845

PubMed Abstract | CrossRef Full Text | Google Scholar

163. Li Q, Ma Z, Liu Y, Kan X, Wang C, Su B, et al. Low doses of paclitaxel enhance liver metastasis of breast cancer cells in the mouse model. FEBS J (2016) 283(15):2836–52. doi: 10.1111/febs.13767

PubMed Abstract | CrossRef Full Text | Google Scholar

164. Zenitani M, Nojiri T, Hosoda H, Kimura T, Uehara S, Miyazato M, et al. Chemotherapy can promote liver metastasis by enhancing metastatic niche formation in mice. J Surg Res (2018) 224:50–7. doi: 10.1016/j.jss.2017.11.050

PubMed Abstract | CrossRef Full Text | Google Scholar

165. Thompson KN, Ju JA, Ory EC, Pratt SJP, Lee RM, Mathias TJ, et al. Microtubule disruption reduces metastasis more effectively than primary tumor growth. Breast Cancer Res (2022) 24(1):13. doi: 10.1186/s13058-022-01506-2

PubMed Abstract | CrossRef Full Text | Google Scholar

166. Watanabe K, Yui Y, Sasagawa S, Suzuki K, Kanamori M, Yasuda T, et al. Low-dose eribulin reduces lung metastasis of osteosarcoma in vitro and in vivo. Oncotarget (2019) 10(2):161–74. doi: 10.18632/oncotarget.26536

PubMed Abstract | CrossRef Full Text | Google Scholar

167. Yoshida T, Ozawa Y, Kimura T, Sato Y, Kuznetsov G, Xu S, et al. Eribulin mesilate suppresses experimental metastasis of breast cancer cells by reversing phenotype from epithelial–mesenchymal transition (EMT) to mesenchymal–epithelial transition (MET) states. Br J Cancer (2014) 110(6):1497–505. doi: 10.1038/bjc.2014.80

PubMed Abstract | CrossRef Full Text | Google Scholar

168. O’Shaughnessy J, Kaklamani V, Kalinsky K. Perspectives on the mechanism of action and clinical application of eribulin for metastatic breast cancer. Future Oncol (2019) 15(14):1641–53. doi: 10.2217/fon-2018-0936

PubMed Abstract | CrossRef Full Text | Google Scholar

169. Kopf A, Renkawitz J, Hauschild R, Girkontaite I, Tedford K, Merrin J, et al. Microtubules control cellular shape and coherence in amoeboid migrating cells. J Cell Biol (2020) 219(6):e201907154. doi: 10.1083/jcb.201907154

PubMed Abstract | CrossRef Full Text | Google Scholar

170. Welch MD. Cell migration, freshly squeezed. Cell (2015) 160(4):581–2. doi: 10.1016/j.cell.2015.01.053

PubMed Abstract | CrossRef Full Text | Google Scholar

171. Lämmermann T, Sixt M. Mechanical modes of a’moeboid’ cell migration. Curr Opin Cell Biol (2009) 21(5):636–44. doi: 10.1016/j.ceb.2009.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

172. Tekle YI, Williams JR. Cytoskeletal architecture and its evolutionary significance in amoeboid eukaryotes and their mode of locomotion. R Soc Open Sci (2016) 3(9):160283. doi: 10.1098/rsos.160283

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: microtubules, invasion plasticity, amoeboid, mesenchymal, cancer, 3D migration

Citation: Legátová A, Pelantová M, Rösel D, Brábek J and Škarková A (2023) The emerging role of microtubules in invasion plasticity. Front. Oncol. 13:1118171. doi: 10.3389/fonc.2023.1118171

Received: 07 December 2022; Accepted: 31 January 2023;
Published: 13 February 2023.

Edited by:

Michelle L. Matter, Tulane University, United States

Reviewed by:

Erdem Tabdanov, College of Medicine, The Pennsylvania State University, United States
Christina Stuelten, National Cancer Institute (NIH), United States

Copyright © 2023 Legátová, Pelantová, Rösel, Brábek and Škarková. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Aneta Škarková, aneta.skarkova@natur.cuni.cz

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.