Next Article in Journal
Features of Peripheral Blood Th-Cell Subset Composition and Serum Cytokine Level in Patients with Activity-Driven Ankylosing Spondylitis
Next Article in Special Issue
Cytochalasin B Influences Cytoskeletal Organization and Osteogenic Potential of Human Wharton’s Jelly Mesenchymal Stem Cells
Previous Article in Journal
Can PSMA-Targeting Radiopharmaceuticals Be Useful for Detecting Hepatocellular Carcinoma Using Positron Emission Tomography? An Updated Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cytoskeletal and Cytoskeleton-Associated Proteins: Key Regulators of Cancer Stem Cell Properties

1
Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
2
National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
3
NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, China
4
Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
*
Authors to whom correspondence should be addressed.
These authors contribute equally to this work.
Pharmaceuticals 2022, 15(11), 1369; https://doi.org/10.3390/ph15111369
Submission received: 9 October 2022 / Revised: 2 November 2022 / Accepted: 6 November 2022 / Published: 8 November 2022

Abstract

:
Cancer stem cells (CSCs) are a subpopulation of cancer cells possessing stemness characteristics that are closely associated with tumor proliferation, recurrence and resistance to therapy. Recent studies have shown that different cytoskeletal components and remodeling processes have a profound impact on the behavior of CSCs. In this review, we outline the different cytoskeletal components regulating the properties of CSCs and discuss current and ongoing therapeutic strategies targeting the cytoskeleton. Given the many challenges currently faced in targeted cancer therapy, a deeper comprehension of the molecular events involved in the interaction of the cytoskeleton and CSCs will help us identify more effective therapeutic strategies to eliminate CSCs and ultimately improve patient survival.

1. Introduction

A steady stream of research has led to a degree of understanding of the tumorigenesis and growth of primary tumors and the development of complex and effective treatments that can significantly prolong patient survival. However, an inescapable problem is that tumor recurrence and metastasis remain a major cause of high mortality in patients with cancer, even after radical surgery combined with adjuvant therapy [1]. Moreover, chemotherapy resistance is also a troubling and intractable problem in the course of tumor treatment [1]. However, the effective understanding of both phenomena is still very limited. Theories related to cancer stem cells (CSCs), as a rising star in tumor research, seem to help understand the abovementioned problems to a certain extent. CSC theory suggests that CSCs can drive tumor growth, promote tumor progression, and initiate mechanisms related to distant metastasis and drug resistance, features that ultimately lead to dismal clinical outcomes [2,3,4]. Therefore, the eradication of this specific group seems to be a priority after recognizing that CSCs with these characteristics may be the culprits. Before addressing this issue, a comprehensive understanding of the biological drive, state regulation and maintenance of CSCs is necessary.
Similar to the case of normal stem cells, CSCs are thought to present in a niche [5]. The niche of CSCs is their specific survival microenvironment, which regulates the fate of CSCs through secreted factors and cell–cell contacts [5]. Niches are three-dimensional structures composed of extracellular matrix components, signaling molecules, and other cells [6]. Through the mechanical interaction of their niche, cells are mechanically loaded, resulting in the possible deformation of the cell membrane, cytoskeleton, and nucleus, thus triggering the secretion of relevant signaling molecules into the niche [5,6]. These signals and secretory factors in turn regulate the metabolism, morphology, and mechano-sensitivity of secretory cells [6]. One study reported that the cytoskeleton is closely related to the activity of CSCs [7]. The eukaryotic cytoskeleton, which is made up of microfilaments, intermediate filaments, and microtubules, is a dynamic and intricate three-dimensional network that exists inside the cytoplasm [8]. The main roles of the cytoskeleton include mechanical support, cell shape regulation, the facilitation of cell migration, and intracellular transport [8,9]. It can also provide locations for the localization and binding of signaling molecules as scaffolds for signaling cascades [8,9]. The dysregulation of the cytoskeleton is closely related to a variety of diseases, especially cancer [10]. Different cytoskeletal components and remodeling processes have profound effects on the behavior of CSCs [11]. Moreover, the cytoskeleton can play a role in regulating cellular bioenergetics in CSCs by dynamically controlling the mitochondrial structure and function of CSCs, in addition to affecting the niche of CSCs [12].
It is evident that understanding the influence of the cytoskeleton and related proteins on various biological behaviors of CSCs will help to further solve the clinical treatment challenges of cancer. Therefore, we outline existing research that supports the significance of cytoskeletal components in controlling the structure, bioenergetics, and function of CSCs. A greater understanding of the behavior and regulatory factors of CSCs will help with the creation of innovative treatments for metastatic, drug-resistant malignancies.

2. Cancer Stem Cells

In the early 1990s, CSCs were discovered in leukemia and were isolated by recognizing the expression of their characteristic surface markers, CD34+CD38 [13,14]. Subsequently, CSCs expressing different surface markers were identified in a large number of solid tumors, such as CD133+CXCR4+ CSCs in pancreatic cancer and CD44+CD24 CSCs in breast cancer [15,16]. It was shown that these special cells are also part of the tumor body [17]. CSCs have a strong capacity for self-renewal, that is, the process of generating at least one daughter cell that retains stem cell characteristics by symmetric or asymmetric division [18]. The expansion of CSCs in a symmetrical division leads to unrestricted cell growth, which directly leads to tumor formation [19,20]. CSCs, like regular stem cells, are controlled by the Wnt/β-catenin, Sonic Hedgehog (Hh), and Notch pathways responsible for self-renewal [21,22,23]. Understanding the regulation of CSCs’ self-renewal may provide more options for cancer treatment. Another great feature of CSCs is their capacity to differentiate into various cell types [24]. Under normal circumstances, multiple signaling pathways stably regulate these two important properties of stem cells to form a balance that is conducive to normal proliferation and differentiation [25]. However, when this regulatory balance is disrupted, uncontrolled CSCs grow and migrate in a frantic manner, ultimately leading to tumor progression and metastasis [26].
CSCs are located in niches, specialized anatomical areas within the tumor microenvironment [27]. These unique niches contribute to the maintenance of the aforementioned characteristics of CSCs and promote their phenotypic flexibility while shielding them from the immune system [23,28]. Aberrant tumor proliferation and vascular rarefaction lead to a tumor microenvironment characterized by hypoxia, acidity, and malnutrition [29]. Therefore, CSCs must effectively adapt their cellular bioenergetics to cope with these adverse conditions [30]. An in-depth study revealed that CSCs prefer mitochondrial oxidative metabolism [31]. Cancer cells carry out aerobic glycolytic metabolism, while CSCs mainly rely on oxidative phosphorylation (OXPHOS) [32,33,34]. Subpopulations of cancer cells switch between glycolysis and OXPHOS to meet the energy demands of survival, also embodying metabolic plasticity [35]. Compared with general tumor cells, mitochondrial mass and membrane potential were found to be increased in CSCs, reflecting an enhanced mitochondrial function and increased oxygen consumption rate [36,37,38,39,40]. Additionally, a high mitochondrial mass suggests a stem cell phenotype that is linked to a potential for metastasis and resistance to DNA damage [41]. Furthermore, CSCs are considered to be an important contributor to chemoresistance due to their well-defined quiescent phenotype, endothelial–mesenchymal transformation (EMT), multidrug resistance (MDR), and resistance to DNA damage-induced apoptosis [42,43,44,45].

3. Cytoskeleton of the Cell

The cytoskeleton is mainly composed of three structures: microfilaments made of G-actin and F-actin, microtubules made of α and β-tubulin, and intermediate filaments (IFs) made of different keratins and vimentin [8,46]. Any changes, including those of the cellular structure and the rearrangement and relocation of organelles, may lead to changes in cellular metabolism that enhance cell migration and invasion characteristics [9]. Actin and microtubules in the cytoskeleton play supportive and key regulatory roles in these important cellular processes [47]. The unfolding of the actin network is essential to the majority of cellular processes [48]. The ability of actin to freely switch between polymeric F and monomeric G actin forms confers the fast remodeling of the actin cytoskeleton in response to internal and external stimuli [49]. Moreover, this cytoskeletal remodeling plays a crucial role in cellular integrity, motility, and membrane trafficking [50]. Actin can synthesize slowly growing pointy ends and quickly growing barbs through self-polymerization in vitro, and intracellular polymerization is tightly regulated by actin nucleation and actin-severing proteins [51]. Moreover, actin filaments can generate various pseudopodia that cells might exploit to investigate the extracellular environment during invasion and metastasis [52]. Actin reorganization also occurs during mitochondrial fission [53]. Mitochondria can travel via dendrites and axons along actin filaments [12]. During the actin breakdown phase, fragmented mitochondria rapidly fuse, accelerating mitochondrial integrity repair and maintaining mitochondrial homeostasis [54]. In addition, F-actin cages around dysfunctional mitochondria are triggered to assemble, preventing the proliferation of damaged mitochondria [55]. In conclusion, the actin cytoskeleton is critical for the spatial domain transport, dynamics and quality control of mitochondria. Actin-nucleating agents that stimulate the formation of actin filaments are essential for actin activity. Known actin-nucleating proteins include formins, tandem WASP homology 2 (WH2) nucleators, and the Arp2/3 complex [56]. The ability to polymerize and depolymerize is critical to actin, and these processes are primarily regulated in space and time by the actin-binding protein (ABP) family [57]. In cancer cells, the homeostasis between G and F actin and their association with ABPs is frequently altered, leading to dysregulation [58]. ABPs are categorized as monomer-binding proteins, cross-linking and binding proteins, end-capping and severing proteins, anchoring proteins, signaling proteins, and stabilizing proteins based on their roles [59].

3.1. Monomer-Binding Proteins

Monomer-binding proteins mainly include profilin, twinfilin, and thymosin β4 [60]. Four different types of profilin (PFN) exist (PFN1-4), with PFN1 being extensively expressed in all tissues [61]. PFN catalyzes the exchange of ADP with ATP on G-actin monomers, which is essential for actin polymerization [62]. PFN modulates membrane protrusion by binding to N-WASP and VASP, which also increases the intracellular PFN concentration and enhances the elimination of stinger-terminated actin monomers, resulting in depolymerization [63]. The phosphorylation/dephosphorylation of serine 3 is a major regulator of PFN activity. In addition, in combination with PIP2 and cortactin, intracellular pH also modulates the activity of PFN itself [64]. Twinfilin (TWF), an actin monomer sequestering protein, exists in two isoforms of TWF-1 and -2 in the human body [65]. TWF is mainly found in lamellar pseudopods, localized in subcellular regions with a high actin turnover. By binding to ADP-G actin, TWF prevents G-actin from being added to actin filaments [66]. Thus, TWF mainly regulates actin polymerization/depolymerization by blocking nucleotide exchange on actin monomers [67]. Furthermore, TWF is involved in cell migration and the EMT, and it controls the cell cycle by affecting the mTOR pathway [68,69]. Its synthesis is regulated by Cdc42-downstream signaling and Rho GTPases Rac1 [65]. Thymosin β4 (Tβ4) is involved in cytoskeletal reorganization by buffering intracellular G-actin concentrations [70]. It is itself a significant G-actin sequesterer that activates Cdc42 and Rac by activating various signaling pathways [71,72]. Moreover, many studies have suggested that Tβ4 can activate the hypoxia-inducible factor 1 (HIF-1) gene and participate in processes such as the EMT and angiogenesis through the AKT and Notch/NF-κB pathways [73,74,75].

3.2. Cross-Linking and Bundling Proteins

Members of the cross-linking and bundling protein family include fascin, filamins, spectrins and alpha-actinin [76]. Fascin, required for actin binding and bundling, is present in tissues as three isoforms (fascin-1, -2, and -3) [77]. The special beta trefoil of fascin forms actin-binding sites that bind dozens of parallel actin filaments together to form tight, stiff filamentous pseudopods [78]. These actin bundles act as proprietary channels that deliver signaling molecules from the cell core to the cell leading edge [79]. The Rho family of GTPases is a small (~21 kDa) family of signaling G proteins, of which members RhoA and Rac1 act upstream of fascin via protein kinase C (cPKC) to regulate actin binding [80]. F-actin cross-linking by fascin-1 involves the N-terminal and C-terminal domains of fascin-1, and a major mechanism that inhibits the actin-bundling activity of fascin-1 is the phosphorylation of an N-terminal motif (S39 in human fascin-1) by conventional isoforms of protein kinase C (cPKC) [81]. Rac1 and RhoA inhibit actin binding by promoting the cPKC phosphorylation of S39 [82]. Of the three isoforms of filamin (FLN), FLNA and FLNB are widespread while FLNC is confined to cardiac and skeletal muscle [83]. One of the main roles of FLN is to connect actin filaments to the cell membrane [84]. FLN is a homodimeric protein found in stress fibers, lamellar pseudopods and filamentous pseudopods [48]. The N-terminal domain is an actin-binding region featuring F-actin-, α-actin-, β-spectrin-, and fibrin-binding sites [85]. The C-terminus is a repeating rod region through which protein dimerization occurs in a tail-to-tail manner [86]. Functionally, in addition to helping actin form orthogonal branches, FLNA is involved in linking many receptors related to cell signaling and the cell cycle [87]. FLNA is mainly regulated by the phosphorylation of residue S2152 [88]. Spectrins construct hexagonal lattices beneath the plasma membrane to keep the membrane cytoskeletal network stable [89]. The α- and β-spectrin genes, which are widespread in cells, encode the two isoforms of spectrin [90]. The α and β subunits are arranged head-to-head to form an antiparallel tetramer that constitute the platform for the binding of channel proteins, receptors, and transporters [91]. On the one hand, spectrin participates in cell migration through actin-dependent and non-actin-dependent mechanisms, and it can bind to calcium or calmodulin to participate in cell proliferation [92]. This is due to the fact that at normal calcium levels, spectrin proteins are found along the edges of cells and diffuse throughout the cell as calcium concentrations increase [93]. Spectrin binds to calmodulin-dependent protein kinase II (CaMKII), which activates the PI3K/Akt signaling pathway to promote proliferation [94]. Additionally, spectrin is involved in hypoxia-induced, angiogenesis-mediated cytoskeletal remodeling, a process regulated by c-Jun N-terminal kinase (JNK) signaling [95]. The α actinin (ACTN) that belongs to the spectrin superfamily is present in all cells [96]. ACTN exists in four forms (ACTN 1-4) in the human body and is crucial for the formation and stabilization of stress fibers [96,97]. ACTN links the cytoskeleton with transmembrane proteins, stabilizes the cell structure, and provides a scaffold for the integration of signaling molecules into specific sites [98]. The PIP3 produced by PI3K activation damages the interaction of ACTN with actin and integrins, thereby impairing the structure of focal adhesion and promoting cytoskeletal remodeling [99].

3.3. Anchoring Proteins

Anchoring proteins mainly include the ezrin–radixin–moesin (ERM) family and merlin [100]. Ezrin is mainly expressed by the vil2 gene in epithelial cells [101]. Its C and N domains interact with the integral membrane proteins in the actin cytoskeleton and the plasma membrane, respectively [102]. This allows ezrin to form connections between the actin cytoskeleton and the plasma membrane and to respond to extracellular signals [103]. Ezrin is also closely associated with multiple signaling pathways, including Rho, PI3K, AKT, and MAPK [104]. Moesin, another member of the ERM family, binds to actin via the C-terminus, thereby attaching actin to the plasma membrane [105]. Moesin, localized in filopodia and microvilli, is involved in the EMT, cell adhesion, and membrane fold formation [106,107]. In addition, moesin affects cell division and spindle–actin communication by binding to microtubules [108]. Radixin, encoded by chromosome 11, has a central α-structural domain with an F-actin-binding site at the C-terminus [109]. Radixin is essential for cytoskeletal organization, as well as cell motility and adhesion, as it cross-links to actin on the cell surface [110]. Merlin, encoded by chromosome 22, has two isoforms: isoform 1 and isoform 2 [111]. Lacking a conserved actin-binding site, the N-terminus of merlin is the actin-binding domain [112]. Merlin plays an important role in the intracellular effectors that control cell proliferation and adhesion, as well as linking F-actin and transmembrane receptors [112].

3.4. Capping and Severing Proteins

Gelsolin is a ubiquitous capping and severing protein that interacts with G- and F-type actin to regulate actin polymerization through severing, capping, and nucleation [113]. The gene encoding it produces two isoforms that are localized in the plasma and cytoplasm [114]. Gelsolin directly or indirectly alters lipid signaling by binding to kinases and lipases [115]. Cofilins are evolutionarily conserved capping and severing proteins [116]. Cofilin has binding sites for both F-actin and monomeric G-actin. The binding of cofilin to actin filaments alters the orientation of the subunit, which leads to filament severance, producing barbed ends, the preferred site for Arp2/3 binding [117]. The regulation of cofilin mainly depends on the phosphorylation of the LIMK/TESK kinase at ser-3 or pH, PIP2, or cysteine oxidation [118,119]. Villin is a significant part of the brush border cytoskeleton in differentiated epithelial tissue, where it binds, caps, severs and bundles actin filaments [120]. Villin has three actin-binding sites, two of which retain calcium-dependent activity at the core [121]. Villin maintains an autoinhibitory conformation at normal physiological calcium concentrations, but its structural conformation changes and binds to actin with increasing intracellular calcium concentrations [122]. The phosphorylation of tyrosine residues within the core of villin promotes actin severing and binding in multiple modalities, thereby increasing cytoskeletal fluidity and affecting its mechanical properties, ultimately enhancing cell movement [123]. A study showed that the binding of villin to F-actin is regulated by calcium concentration, PIP2 or tropomyosin [124].

3.5. Stabilizing Protein and Signaling Protein

Tropomodulins are stabilizing proteins that wrap around the growing end of actin to prevent the dissociation or addition of G-actin [125]. In addition, tropomodulins can regulate actin dynamics by acting as actin-nucleating agents [125]. Furthermore, tropomodulins regulate actin filament assembly, stability and length via capping [126]. The nucleotide concentration of actin affects tropomodulins’ affinity for G-actin monomers [127]. Ena/VASP is a signaling protein that is essential in the formation and elongation of filamentous pseudopods [128]. The C-terminus of this signaling protein has binding sites for G and F actin, and the protein itself promotes tetramerization, which is significant for actin extension [129]. Ena/VASP proteins are conserved regulators of actin dynamics and play important roles in a variety of physiological processes including morphogenesis, axon guidance, endothelial barrier function, and cancer cell invasion and metastasis [130]. The anti-capping model of Ena/VASP function appears to be the simplest explanation for many of the known cellular biological and biochemical properties of this protein family. Another biochemical property of Ena/VASP proteins is their ability to nucleate actin filaments in vitro, but the importance of this effect in vivo remains to be confirmed [130]. The interaction of Ena/VASP with actin and other proteins can be affected by the phosphorylation of PKA and the dephosphorylation of protein phosphatases [131] (Figure 1).

3.6. Microtubule-Associated Proteins

Microtubules are one of the fundamental constituents of the cytoskeleton and are composed of α- and β-tubulin heterodimers bound together [132]. Microtubules play critical roles in cell morphology, cell division, vesicle transport and cell signaling [132]. Microtubule-associated proteins (MAPs) bind to microtubules, connect them to other organelles, bind them, and transport related substances [133]. Tau is a common MAP that manages microtubule polymerization and stability to govern microtubule protein dynamics [134]. Excessive phosphorylation leads to a decreased affinity between Tau and microtubules, which alters post-translational modifications and destabilizes the cytoskeleton, ultimately resulting in a diminished EMT and invasiveness [135]. Microtubule-associated protein 2 (MAP2) stabilizes microtubule growth by cross-linking microtubules to intermediate filaments, leading to microtubule stiffness activation [136]. Katanin is a heterodimeric protein composed of katanin P60 and katanin P80 subunits that exerts its microtubule-cutting function by deploying ATP hydrolysis to extract microtubulin dimers at the lattice and break down the polymer [137]. In addition, angiotensin II receptor-interacting protein 3 (ATIP3), one of the structural MAPs localized along the microtubule lattice, is an effective microtubule stabilizer that binds to end-binding proteins (such as EB1) in the cytoplasmic lysate, thereby attenuating microtubule dynamics [138]. The four abovementioned MAPs belong to microtubule lattice-binding proteins, which are localized along the length of microtubules.
Microtubule motility proteins include kinesins and dyneins that transport molecules along microtubule tracks [139]. Kinesins consist of different isoforms of family proteins that are involved in individual cellular activities. Classical kinesin with an N-terminal motility domain and kinesin family member 14 (KIF14) with an intermediate motility domain deploy ATP hydrolysis to generate kinesin motility with mechanical force toward the plus-ends of growing microtubules [140,141]. Dyneins transport in the opposite direction to kinesins, moving toward the minus-end of microtubules and transporting intracellular cargo from the cell periphery to the center in a retrograde direction [142]. Kinesins and dyneins play important roles in different microtubule-dependent activities, intracellular vesicle transport, organelle transport, and mitotic spindle organization [143]. Stathmin (STMN1) is one of the most prominent microtubule destabilizers, reducing the length of microtubule polymers by indirectly binding microtubule protein subunits in a bent form, thus promoting depolymerization [144]. In addition, STMN1 induces microtubule instability by specifically interfering with the lateral binding of microtubule protein subunits at the microtubule ends, acting at both the plus- and minus-ends [145]. In addition, MAPs that preferentially contain polymerized microtubule plus-ends are referred to as plus-end tracking proteins (+TIPs) [146]. End-binding proteins (EBs) are typical +TIP protein types, including EB1, EB2 and EB3, which precisely bind to the plus-end of microtubules and associate with stable GTP caps for microtubule growth [147]. Cytoplasmic linker-associated proteins (CLASPs) are a conserved class of +TIPs proteins that contribute to microtubule stabilization [148]. Members of the calmodulin-regulated spectrin-associated protein (CAMSAP) family, consisting of CAMSAP1, CAMSAP2 and CAMSAP3, have recently been described as microtubule minus-end-binding proteins [149]. They may regulate the stability and localization of microtubule negative ends, thereby organizing non-centrosomal microtubule networks sufficient for cell division, migration and polarity [150] (Figure 2).

3.7. Other Components of the Cytoskeleton

The myosin superfamily includes myosins with different structures and functions encoded by dozens of genes [151]. The main role of myosin is to convert chemical signals into mechanical forces, a process that is achieved by its sliding along actin filaments involved in ATP hydrolysis [152]. A variety of important cellular functions such as intracellular signal transduction, cell migration and tumor suppression can be seen with traces of myosin involvement [153]. IFs are dynamic, nonpolar fibrillar structures highly concentrated in desmosomes and hemidesmosomes [154]. IFs share a common structure: an N-terminal head domain, a C-terminal tail domain, and a central rod domain [155]. IFs undergo dramatic structural changes upon the receipt of relevant signals, providing structural support and participating in the control of processes such as cellular proliferation and apoptosis [156]. The regulation of IFs’ organization mainly relies on the interaction of IFs with other proteins, the phosphorylation of some signaling pathways, or post-translational modifications [157]. According to different structures and localizations, intermediate filament proteins can be divided into: type I and type II-acidic and basic cytokeratins; type III-vimentin, glial fibrillary acidic protein, desmin, synchronization protein, and peripheral protein; type IV-neurofilament and α-internexin; type V-lamins; and type VI-synemin and nestin [158]. Keratin binds to integrins via plectin to stabilize hemidesmosomes, causing cell migration and adhesion to be stabilized [159]. Vimentin, which is abundantly expressed in normal mesenchymal cells, is primarily responsible for cellular integrity and stress tolerance [160].

4. Cytoskeleton and CSCs

CSCs are generally considered to be the main culprit for cancer metastasis and chemotherapy resistance. The different components of the cytoskeleton, remodeling processes, and interactions with CSCs enable CSCs to adapt to unique tumor microenvironments, thus maintaining cell stemness and migratory activity.

4.1. Actin and CSCs

Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are important signaling molecules that regulate drug resistance and cancer stem cell biomechanics. [161]. YAP/TAZ proteins act as mechanosensors in response to physical stimuli involving the actin cytoskeleton [162]. YAP/TAZ proteins have been shown to play a two-sided role in the Wnt signaling pathway, which is critical for intercellular function and self-renewal capacity [163]. This is mainly because YAP/TAZ proteins are components of the β-catenin destruction complex that translocates to the nucleus upon the activation of the Wnt pathway [162]. An increased extracellular matrix (ECM) stiffness activates the YAP/TAZ-downstream Rho/ROCK pathway, which facilitates the survival of CSCs [164]. The activation of integrin and focal adhesion kinase (FAK) contributes to focal adhesion formation, leading to the activation of Rho-GTPase and stress fibrillogenesis. Meanwhile, focal adhesions require actin polymerization. These together lead to the repression of YAP/TAZ transcription factors, resulting in negative effects on CSCs [165]. In this case, myosin increases the tension on the actin network after a cell has spread, while F-actin reduces tension by dissociating to maintain tension balance [164].

4.2. Monomer-Binding Proteins and CSCs

PFN is essential for cell motility in vivo through the regulation of actin polymerization kinetics [166]. Cell migration and intercellular adhesion can be inhibited by reducing the expression of PFN in cancer cells [93]. In colorectal cancer, changes in the invasive, migratory and self-renewal abilities of HT29 CSCs were found to be consistent with the rise and fall of PFN2 expression levels. Furthermore, PFN2 directly regulates the expression of EMT markers (E-cadherin) and stemness markers (SOX2, CD133 and β-catenin) [167]. SOX2 is a transcription factor that is essential for the regenerative capacity of stem cells, as well as for the maintenance of pluripotency [168]. Thus, PFN2 plays an important role in the stemness and metastatic potential of CSCs by regulating related transcription factors. Additionally, the knockdown of PFN1 in breast cancer cells was shown to result in the diminished expression of CSC-related genes, further demonstrating the important regulatory role of PFN on CSC-related properties [169]. Tβ4 relies on the cytoskeletal organization of actin to exert a regulatory role on tumorigenicity and metastatic capacity in mouse fibrosarcoma cells [170]. Tβ4 is overexpressed in a variety of tumors and maintains the cell stemness of CSCs by increasing the EMT [171]. Because of its important role in promoting the tumorigenic properties of colorectal CSCs, Tβ4 may have important implications for therapeutic intervention in human colon cancer [172]. Moreover, the expression of Tβ4 is closely associated with the expression of the CSC marker CD133 in gastric and ovarian cancers, thus having an impact on tumor metastasis [173]. In pancreatic cancer, Tβ4 mainly regulates CSCs by activating the JNK pathway and promoting the expression of pro-inflammatory cytokines, thereby promoting cancer progression [174]. This may be due to the fact that Tβ4 first enhances the bone morphogenetic protein (BMP) pathway, which activates JNK through the TAB1 and TAK1 complex [175,176]. Of course, further studies are needed to elucidate the exact pathway of Tβ4-induced JNK activation. Additionally, the increased expression of Tβ4 promotes the migration and metastasis of CSCs, mainly through the activation of Rac and the elevation of the IQGAP1/ILK complex [177]. TWF, a conserved actin-binding protein, is also a prime candidate target for the downregulation list of miR-206 [178]. It has been reported that hsa-miR-206 attenuates the stemness and metastatic ability of breast CSCs by reducing their self-renewal and invasive ability. TWF1 could rescue the invasive phenotype of miR-206 by enhancing the activity of the mesenchymal lineage transcription factor-megakaryocytic leukemia 1 (MKL1) and actin cytoskeleton dynamics [178]. On the other hand, a systemic RNA interference screening study revealed a strong association of TWF1 with chemosensitivity and cell motility [179].

4.3. Cross-Linking and Bundling Proteins Interact with CSCs

Fascin, as an actin-binding protein, directly mediates chemoresistance in breast cancer by activating FAK [180]. Moreover, fascin activates β-catenin signaling and promotes breast CSC function, mainly through focal adhesion kinase (FAK) [181]. The upregulation of fascin expression results in cytoskeletal changes that promote metastasis [182]. After the knockdown of fascin in ovarian cancer stromal cells with high fascin expression, we found that CSC activity, metastasis, and the EMT were reduced through pathways such as Rac1, RhoA, and NF-κB [183]. As a result, the increased expression of fascin in most aggressive cancers often represents the possibility of metastasis. FLNA is able to remodel the actin cytoskeleton of CSCs, leading to enhanced tumor metastasis [184]. It interacts with Rho GTPases, which activate cell migration, and Ras GTPases, which inhibit cell migration, to promote metastasis [184]. The downregulation of FLNA increases the destruction of single- and double-stranded DNA in tumor cells after cisplatin therapy, increasing chemosensitivity [185]. Moreover, a lack of FLNA arrests the cell cycle in the G2/M phase and increases angiogenesis by promoting the expression of VEGF [186]. In head and neck CSCs, the activation of CD44 alters FLN expression, resulting in enhanced cell migration and chemoresistance [187]. According to research, spectrins may be closely associated with tumorigenesis, progression and metastatic processes [188]. Spectrin is highly expressed in early-stage colorectal cancer but lower in advanced or metastatic cells [189]. It was shown that colorectal cancer cell viability and cell contacts were reduced and metastasis was increased when spectrin was knocked out. Furthermore, a marked decrease in spectrin expression may result in the loss of DNA mismatch repair proteins [190]. In addition, β2 spectrin was shown to inhibit the properties of hepatic CSCs through β-catenin-induced differentiation, which is a new strategy for hepatocellular carcinoma prevention and differentiation therapy [191]. ACTN is involved in cell differentiation and cancer metastasis by modulating the activity of several signaling pathways and recombinant actin filaments [93]. It was shown that the potential mechanism of the ACTN4-mediated properties of CSCs mainly involves the Akt/GSK-3β/β-catenin axis. The ACTN4-mediated stabilization of β-catenin is closely related to Akt/GSK-3β signaling. ACTN4 promotes the EMT and cell cycle progression by stabilizing β-catenin, maintaining the properties of CSCs, and leading to drug resistance [192,193]. Furthermore, studies have shown that high levels of ACTN4 expression are related to malignancy, metastasis, poor prognosis, and chemotherapy resistance in numerous tumors, including pancreatic, ovarian, and bladder cancers [93,194].

4.4. Anchoring Proteins and CSCs

The increased expression of ezrin leads to increased malignancy and decreased survival in aggressive cancers [195]. Ezrin tends to be expressed more on the apical surface of tissues in non-invasive tumors, whereas in invasive cell lines, it tends to be expressed in local membrane folds and filopodia, which are more favorable for promoting metastasis [196]. Furthermore, the ectopic expression of phosphomimetic forms of ezrin promotes cancer progression and metastasis in vitro and in vivo [197]. Ezrin and CD44 are co-highly expressed in breast CSCs, which is associated not only with poor prognosis but also with the resistance of CSCs to chemotherapy [198]. In pancreatic ductal adenocarcinoma (PDAC), the level of ezrin in CSCs is significantly higher than that in normal cancer cells. The severe impairment of CSC frequency, self-renewal capacity, and tumor initiation potential was observed following the knockout or inhibition of ezrin. These all suggested that ezrin affects the properties of CSCs in PDAC [199]. Ezrin is associated with defective adhesion turnover and a loss of directional migration, leading to tumor invasion and metastasis [200]. The potential mechanism could be as follows. On the one hand, ezrin connects the cytoplasmic tail of CD44 to F-actin, leading to cytoskeletal remodeling, and changes in actin cytoskeletal dynamics and cell shape could guide stem cell differentiation [201]. On the other hand, ezrin can also maintain CSC properties by regulating actin polymerization through ROCK inhibition [199]. Moesin, like other ERM proteins, has also been implicated in cancer progression [202]. Moesin is commonly overexpressed in high-grade glioblastoma, and its mode of action correlates with the CSC marker CD44. The main mechanism of action of moesin is to increase the expression of CD44 in the Wnt/β-catenin signaling pathway and to enhance the positive feedback effect on this pathway. Furthermore, moesin was shown to increase the expression of SOX2, promoting the functional transition of glioblastoma to an aggressive stem cell phenotype [203]. Merlin is encoded by the tumor suppressor gene NF2 [204]. This protein regulates YAP/TAZ proteins through the merlin/NF2/YAP/TAZ axis [205]. YAP/TAZ proteins are key regulators of the properties of breast cancer CSCs [206].

4.5. Capping and Severing Proteins Interact with CSCs

Gelsolin is closely associated with properties such as oncogenic phenotype, the EMT, cell motility, apoptosis, proliferation and differentiation [207]. Furthermore, gelsolin interferes with TGF-β1-driven CSC differentiation through the EMT process in breast cancer cells [208]. Gelsolin affects the differentiation and properties of stem cells by regulating stem cell-related transcription factors such as Nanog, SOX2, and OCT4 [208]. Chemotherapeutic drugs induce hepatocellular carcinoma cell death by activating cofilin-1, a process associated with the interaction of Bcl-2-associated X protein and ROS accumulation. Thus, the phosphorylation of cofilin-1 leads to chemoresistance [209]. High levels of cofilin-1 have been shown to be prognostic biomarkers and predictors of drug resistance [93]. The overexpression of cofilin in prostate cancer leads to an enhanced EMT and promotes metastasis and CSC properties [210]. Studies have reported that the knockdown of villin in specific cell lines using siRNA resulted in cell growth arrest, demonstrating its importance in carcinogenesis [211]. Villin can also be used as a marker of gastric CSCs and a biomarker of metastatic lung adenocarcinoma [212,213].

4.6. Stabilizing and Signaling Proteins Interact with CSCs

Highly expressed tropomodulins in hepatocellular carcinoma lead to increased invasiveness, metastasis, CSC properties, and matrix metalloproteinase (MMP) expression through the activation of the PI3K–AKT signaling pathway [214]. Tropomodulins increase the expression of MMP-13 and NF-κB in breast cancer, which contributes to enhanced tumor invasion, stemness and metastasis [215]. VASP is involved in ECM-mediated β1-integrin-FAK–YAP/TAZ signaling, which is closely related to the regulation of CSC properties [216]. Furthermore, the increased expression of Ena/VASP in PDAC and colorectal cancer (CRC) were found to be significantly associated with liver metastasis and lower survival [216]. In gastric cancer cells, the expression of VASP can be inhibited by miR-4455, thereby reducing VASP-mediated properties such as proliferation, migration, stemness and invasion [217].

4.7. Microtubule-Associated Proteins Interact with CSCs

Tubulin regulates the EMT and contributes to the formation of lamellar and filopodia, promoting cancer cell stemness and metastasis [218]. The ectopic expression of Snail or Twist facilitates α-tubulin decarboxylation and microtubulin-based microtentacle formation, which aid in invasion and migration [219]. Tau can modulate cell cycle processes and related signaling pathways in cancer to affect stem cell-like phenotypes [93]. For example, Tau activates the MAPK pathway involved in prostate cancer progression by binding to PI3K [220]. The high expression of tau mRNA in breast cancer often indicates chemoresistance [221]. It was confirmed that katanin contributes to the formation of CSCs, leading to metastasis [222]. The main principle may be that katanin acts as a microtubule-severing protein that cleaves cellular microtubules into short pieces and activates JNK [223]. In addition, upregulated katanin may increase microtubule dynamics, accelerate the cell cycle, and increase cell viability and cell migration, thereby promoting tumor metastasis [224]. A previous study suggested the involvement of microtubule stabilizer ATIP3 in the inhibition of ERK1/2 activity. ATIP3 leads to the inhibition of CSCs and the EMT through ATIP3/ERK1/2-Snai2 signaling, reducing cell proliferation, migration and invasion [225]. Among kinesins, KIF11 was found to enhance the stemness of cancer cells by promoting the expression of stemness transcription factors (NANOG and OCT4), leading to cell proliferation and resistance to chemotherapeutic agents [226]. STMN1 leads to microtubule depolymerization, which promotes the activation of Rho, thereby enhancing the EMT and cell stemness [227]. Moreover, microtubule disruption promotes the assembly of adherent spots and enhances cell migration [228]. CAMSAP3 protects lung cancer cells from the EMT by inhibiting Akt activity through microtubule regulation, whereas CAMSAP3 deficiency promotes the EMT and stemness maintenance in these cells [229].

4.8. Other Components of the Cytoskeleton on CSCs

The nuclear transfer of cells through the dense extracellular matrix is one of the most important steps in the process of cancer metastasis [230]. Thus, nuclear translocation is considered to be a key limiting factor for the efficient spatial migration of cancer cells [230]. It was demonstrated that myosin IIB enhances the ability of nuclear translocation in breast CSCs, thereby enhancing stem cell invasiveness [231]. Myosin IIB combines a nuclear scaffold structure with the actin cytoskeleton to facilitate the extrusion of nuclei through narrow spaces, resulting in effective 3D collagen invasion [232]. In addition, myosin IIA is involved in promoting the EMT, and the transition between myosin IIB and myosin IIC is critical for the EMT, contributing to stemness maintenance by influencing cell contractility [233,234]. Vimentin, a type III IF, is one of the key biomarkers for the EMT and is usually upregulated during cancer metastasis [235]. Vimentin regulates EMT-related genes, including Twist, Snail, ZEB1/2, and Slug, as well as key epigenetic factors [236]. Moreover, it relies on inducing genes associated with self-renewal to inhibit cell differentiation and to upregulate their pluripotent potential, thereby increasing the stemness of CSCs and promoting tumor metastasis and chemoresistance [237]. Nestin is closely related to self-renewal capacity and is considered a stem marker for neurogenic tumors and epithelial or mesenchymal tumors [238]. Nestin may be a useful biomarker and a new target for inhibiting tumor angiogenesis due to its more widespread expression in the proliferating vessels of PDAC [239]. High levels of nestin expression in breast cancer patients are correlated with the upregulation of VEGF, cancer stem cell markers, and proteins that activate Wnt/β-catenin to initiate proliferation [240]. Several keratins (KRT6, 14, 16, and 17) have been reported to be involved in the regulation of different types of cancer stem cells [241]. The interkeratin fusion between KRT6 and KRT14 promotes CSC-related properties in oral squamous cell carcinoma [241]. KRT16 can promote cancer drug resistance and stemness by interacting with the β5-integrin/c-Met signaling pathway [242]. KRT17 regulates stemness and chemoresistance by binding to β4-integrin/FAK, Src, or β-catenin [243] (Table 1).

4.9. Mitochondria-Cytoskeleton Interactions and CSCs

CSCs exhibit elevated mitochondrial fusion, and their metabolism relies on a rearranged cytoskeletal network and OXPHOS [12]. Increased mitochondrial fusion encourages ATP synthesis by OXPHOS, addressing the energy limitation problem for CSC survival [244]. In addition to functioning as a crucial metabolic enzyme in glycolysis, aldolase also interacts with cytoskeletal elements that regulate actin polymerization [245]. Through cytoskeletal rearrangements leading to the spatial redistribution of aldolase, PI3K plays an AKT-independent role in altering glycolysis, thereby increasing energy metabolism [12]. Cytoskeletal rearrangements or regulatory mechanisms between cellular bioenergetics and cytoskeletal regulators are critical for understanding the responses of cancer cells, especially CSCs, to different stimuli [12]. The EMT program has been identified as one of the key regulators of the CSC phenotype [45]. The EMT is also regulated by cytoskeleton–mitochondrial interactions [12]. The EMT is determined in part by the morphological reprogramming of cellular architecture and sustained by a reconstituted cytoskeleton [246]. The aggregation of mitochondria near the cell membrane is essential to facilitate the formation of cytomotor structures such as pseudopods during the EMT [247]. Studies have suggested that ROS may participate in the regulation of the EMT through actin reorganization [12,248].

5. Therapeutic Strategies Targeting Cytoskeleton

5.1. Therapeutic Strategies Targeting Actin

The cytoskeleton is essential for the invasion and migration of cancer cells, making it a promising therapeutic target. Although the concept of cancer therapy targeting actin is not new, other healthy cells may be affected because of severe off-target effects [249]. Therefore, the issue of the clinical application of actin-targeted therapy remains a pressing challenge. At present, a variety of actin toxins and inhibitors, including phalloidin, cytochalasins, jasplakinolide, latrunculins, wiskostatin, CK-666, and CK-869, are widely used for research [250,251,252]. However, with severe and widespread cytotoxicity, the clinical application of these drugs has not been implemented. Thus, it is crucial to develop actin inhibitors with strong specificity and high safety to treat cancer cells. Considering the specificity of actin, researchers have focused on actin-nucleating agents. The use of small-molecule-targeted formalin has been shown to be potentially beneficial for cancer treatment. The small-molecule inhibitor SMIFH2 inhibits formalin activity, and SMIFH2 binds to the FH2 structural domain of actin nucleation, inhibiting actin nucleation and elongation [253]. The use of SMIFH2 was found to enhance the sensitivity of ovarian cancer cells to cisplatin or paclitaxel [254]. Rho expression tends to be increased in tumors, and actin dynamic function in cancer cells correlates with Rho activity [255]. Therefore, the inhibition of Rho or upstream signaling regulators of Rho can block abnormal cytoskeletal activity and may be a promising strategy for cancer therapy [256]. Cdc42 is a small GTPase that activates a variety of downstream effector molecules, including actin-related proteins, kinases, and phospholipases [257]. The Food and Drug Administration (FDA)-approved analgesic drug R-ketorolac (Toradol) inhibits Cdc42 and Rac1 in ovarian cancer cells and is currently in clinical trials (NCT02470299) [258]. Another effective drug, MBQ-167, is an inhibitor of Rac/Cdc42 both in vivo and in vitro [259]. It has been shown to suppress the motility viability and clumping of breast cancer cells, but further optimization and development are required before clinical trials [256]. Rac is often overexpressed or overactivated in a variety of cancers and is also a small GTPase protein [260]. The drugs currently being developed to inhibit Rac activity have only been tested at the cancer cell level, but none appear to be entering clinical trials [259]. ROCK is another target proposed in multiple studies [256]. ROCK1/2 serine/threonine kinases modulate cell morphology, as well as actin cytoskeleton reorganization, by phosphorylating various ABPs, such as ERM proteins [261]. The inhibition of ROCK1/2 could theoretically disrupt the cytoskeletal dynamics of cancer cell actin and provide therapeutic benefits [262]. However, another study suggested that ROCK inhibition may activate an alternative pathway leading to a more aggressive migratory phenotype [263]. Additionally, currently used ROCK inhibitors lack selectivity for ROCK1 and ROCK2, which have distinct roles in the regulation of cytoskeletal networks [264]. The use of non-selective inhibitors may actually have a promoting effect on certain malignancies and the tumor microenvironment [265]. The combination of novel selective ROCK inhibitors with different anticancer drugs for cancer treatment is anticipated and exciting.

5.2. Therapeutic Strategies Targeting ABPs

Profilin has the potential to be a therapeutic target against a variety of cancers because of its role in cytoskeletal regulation and its location in cancer signaling cascades [266]. Small molecule screens have identified two small molecules (C1 and C2) that prevent profilin from interacting with actin monomers [267]. However, its mechanism and safety need to be further studied before clinical trials. Tβ4 also has potential therapeutic effects on cancer. Silencing the Tβ4 gene in non-small cell lung cancer was found to inhibit tumor progression, suggesting that Tβ4 could be a candidate target for therapy [268]. Tβ4 tends to be highly expressed in rectal CSCs. Interestingly, when using lentivirus to reduce Tβ4 levels in rectal cancer stem cells, this treatment significantly reduced tumor size and aggressiveness in mice [172]. A few newly synthesized drugs have been detected to act as active fascin inhibitors to treat cancer [269]. Compound G2 was found to inhibit fascin-1-directed actin remodeling, an action that caused the destruction of filamentous pseudopods and minimized the migratory and invasive characteristics of colorectal cancer cells both in vitro and in vivo [270]. However, the side effects of this treatment are not fully understood and need to be further studied. Additionally, compounds 3 and 14 were found to substantially downregulate fascin-1 and abolish the EMT, leading to a reduction in the invasiveness and metastatic ability of cancer cells [271]. However, further research is needed to study how these compounds work in vivo and to address any side effects as much as possible. Among them, the small-molecule fascin inhibitor NP-G2-044 has been shown to block tumor invasion and metastasis. This orally available inhibitor binds to fascin and blocks the interaction of fascin with actin filaments [272]. A multicenter Phase 1A clinical trial was designed to evaluate the safety and tolerability of NP-G2-044 in a single daily oral dose for the treatment of patients with refractory solid tumor malignancies (NCT03199586). Overall, the results showed the good absorption and distribution of NP-G2-044 in humans, with initial signals of antitumor and antimetastatic activity observed and no drug-related serious adverse events, dose-limiting toxicities, or patient deaths [273]. Raltegravir is a human immunodeficiency virus 1 integrase inhibitor that disrupts cell motility by directly acting on fascin-1 to cause the breakdown of the actin cytoskeleton [274]. However, its safety and efficacy in humans remain to be tested. Salinomycin was identified as an inhibitor of fascin-1, an ion carrier and antibiotic in its own right. Salinomycin relocates fascin-1 from filamentous pseudopods in PDAC cells, disrupting actin cytoskeleton remodeling and inhibiting cancer metastasis to secondary sites [275]. The antidepressant imipramine has also been identified as a novel fascin-1 inhibitor that significantly reduces fascin-1 expression and disrupts filamentous pseudopod formation and cytoskeletal remodeling [276]. Clinical trials on imipramine are currently underway in ER+/triple-negative breast cancer (NCT03122444) and recurrent glioblastoma (NCT04863950) [269].
The inhibition of ezrin phosphorylation may be an effective strategy for cancer treatment. Researchers screened a small-molecule library of multiple compounds that might interact with ezrin by ion resonance (SPR) technology, and two of these compounds—NSC668394 and NSC305787—were found to have a strong binding affinity for ezrin [277]. They can significantly inhibit the phosphorylation of ezrin, inhibit the interaction between ezrin and F-actin, and achieve the inhibition of oncogenic activities including osteosarcoma cell invasion, migration, and lung metastasis [278]. Furthermore, a study found that NSC668394 in combination with lapatinib, a drug targeting HER2 and EGFR, enhanced the induction of apoptosis and the inhibition of breast cancer cell proliferation [279]. Ezrin is essential for cancer progression, acting as a scaffolding protein and interacting with related proteins in cancer cells [197]. Hence, designing inhibitors to interfere with the interaction of ezrin with related proteins may be another strategy for cancer therapy. For example, small molecules that interfere with the ezrin–L1CAM interaction may be promising therapeutic agents for colorectal cancer [280]. Furthermore, the treatment of ERMs with cytochalasin B was shown to remarkably suppress the metastasis and phagocytic activity of melanoma cells, indicating that the inhibition of actin assembly by ezrin inhibitors may be a potential therapeutic tool for melanoma [281]. Another study found that G1749-A1771 siRNA targeting ezrin mRNA effectively downregulated the expression of ezrin, contributing to the induction of apoptosis and the inhibition of cell proliferation in osteosarcoma cells [282]. In addition, AKT inhibitors (MK2206) or PI3K inhibitors (LY294002) can block ezrin-mediated tumor growth and metastasis by inhibiting the PI3K/AKT signaling pathway [197]. The multi-kinase inhibitor sorafenib (BAY43-9006) promoted apoptosis by inhibiting the ezrin pathway and inhibited angiogenesis and metastasis in a mouse model of osteosarcoma [283]. There are also many cancer drugs targeting ezrin in natural compounds. Recent studies have shown that baicalin exerts antitumor effects by inducing apoptosis and inhibiting cell proliferation and invasion by inhibiting the expression of ezrin [284]. In addition, the binding of celastrol to ROCK2 inhibits the migration of hepatocellular carcinoma, mainly through the impaired ROCK2-mediated phosphorylation of ezrin, resulting in ineffective ezrin activation [285].

5.3. Therapeutic Strategies Targeting Microtubules and IFs

Considering the important role of microtubules in the cytoskeleton, drugs targeting microtubule dynamics are one of the most effective treatments [286]. For example, one of the first compounds to target the cytoskeleton to treat cancer was paclitaxel (PTX), which stabilizes microtubules and effectively prevents cell division in a wide range of cancers, including lung, ovarian, and breast cancers [287]. However, the effectiveness of PTX is limited by various side effects. The main side effects of PTX are allergy and neuropathy. PTX hypersensitivity reactions are usually seen within the first ten minutes of administration and include dyspnea, bronchospasm, urticaria, abdominal pain, fever, or chills, which usually result in the immediate discontinuation of therapy [288]. In addition, the cardiotoxicity caused by PTX administration cannot be ignored. Effective inhibitors of microtubule dynamics also include periwinkle alkaloids, which is widely used in cancer therapy [12]. Given the importance of intermediate filament proteins in various tumor activities, treatment targeting intermediate filaments and their associated signaling networks may also be a promising therapeutic strategy [289]. The naturally derived bioactive compound withaferin-A targets and induces vimentin cleavage and inhibits tumor progression and metastasis in mouse models [290]. Although withaferin-A is currently the only small molecule that inhibits the structure and function of intermediate filaments, it also acts on several other cellular components and lacks specificity [291]. Therefore, there is an urgent need to develop inhibitors that exclusively target intermediate filament proteins to modulate their function, which will be very important for the clinical treatment of cancer patients.

5.4. Therapeutic Strategies Targeting MAPs

The expression of MAPs can greatly influence the efficacy of microtubule-targeted therapy [292]. The MAP tau was identified as a predictive marker for a pathological complete response to paclitaxel in breast cancer patients. Low levels of tau protein expression make mitosis and cytoskeletal microtubules more sensitive to paclitaxel disruption [293]. After long-term treatment, drug resistance has become a major problem in targeting microtubule therapy. These resistance mechanisms are associated with alterations in the microtubule proteins themselves, including alterations in microtubule protein isoform expression, post-translational modifications of microtubule proteins, and the acquisition of microtubule protein mutations [294]. The purine-type compound 5a affects the structure of microtubules and causes apoptosis in cancer cells by targeting the microtubule cleavage protein katanin. This pharmacological effect may bypass the primary resistance mechanism described above. Thus, 5a and its analogs may be new therapeutic options for targeting katanin [223]. KIF11 is currently the most well-studied kinesin in the clinical setting. Ispinesib, a quinazolinone derivative, is the first KIF11 inhibitor to be studied in clinical trials [295]. In a phase I trial (NCT00089973) evaluating the safety and efficacy of ispinesib in breast cancer, antitumor activity was detected in 20% of patients and stable disease was noted in 73% of patients, with 27% having stable disease for 90 days or longer. The most common adverse events reported were neutropenia, elevated liver transaminases, and diarrhea [296]. SB743921 is a recently discovered inhibitor of KIF11. In a clinical trial (NCT00136513) of SP743921 in patients with advanced solid tumors, the drug showed encouraging efficacy without serious toxicity [297]. Other KIF11 inhibitors that have shown promise in cancer therapy include curcumin and various tetrahydro-β-carboline-acetonide hybrids and thione derivatives. Curcumin is a non-specific plant polyphenol extracted from turmeric that exhibits antioxidant, anti-inflammatory, antibacterial and antiviral biological activities [298]. Based on extensive studies, stathmin has recently emerged as a promising drug candidate for the treatment of solid malignancies. To reduce stathmin transcripts in vitro and in vivo and to explore therapeutic approaches against stathmin, a range of specific anti-stathmin agents are being developed [299]. Anti-stathmin nuclease gene delivery via adenovirus reduces multiplication and clonality in breast cancer cells with and without estrogen receptors [300]. The stathmin promoter-driven Aurora-A shRNA adenoviral pathway can be used to manage breast cancer as a complementary tumor-specific therapy [301].

5.5. Therapeutic Strategies Targeting the Metabolism of CSCs

Mitochondrial fusion is important for the energy metabolism of CSCs, so the inhibition of mitochondrial fusion may be a candidate cancer therapeutic strategy. Changes in mitochondrial fusion proteins affect mitochondrial morphology and integrity, making these proteins ideal therapeutic targets [302]. β II protein kinase C (βIIPKC), a selective inhibitor of mitochondrial fusion proteins, phosphorylates fusion proteins and causes the partial loss of GTPase activity, resulting in the fragmentation and dysfunction of mitochondria [303]. OXPHOS inhibitors are expected to be used for the targeted therapy of OXPHOS-dependent tumors. Metformin is one of the most promising inhibitors of OXPHOS, a diabetes agent for cancer therapy [304]. Benzformin is an alternative to metformin that has the advantage of a greater affinity for mitochondrial membranes and easier transport in cancer cells [305]. A novel metformin derivative (IM156) primarily acts on slowly circulating tumor cells that have the ability to evade conventional chemotherapy [306]. These therapies have been shown to inhibit mitochondrial function and CSC survival, but consideration ought to be given to whether the suppression of OXPHOS or a specific component induces an alternative pathway that affects ultimate anticancer efficacy (Table 2).

6. Conclusions

In this review, we summarize the mechanisms by which interactions between the cytoskeleton, cytoskeleton-associated proteins and CSCs lead to tumor metastasis and drug resistance. The effects of various cytoskeletal components, including ABPs and MAPs, and cytoskeletal reorganization on CSCs are emphatically expounded. Additionally, the main metabolic mode of CSCs, OXPHOS, is also modulated by cytoskeletal–mitochondrial interactions. Thus, a detailed understanding of the interactions between the CSCs and cytoskeleton facilitates the development of new cancer treatment strategies to provide better therapy for metastatic and drug-resistant patients.

Author Contributions

Conceptualization, D.W., H.G. and X.G.; investigation, Y.L., Y.C. and C.G.; writing—original draft preparation, D.W. and H.G.; writing—review and editing, D.W., X.G., Y.C. and Y.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Natural Science Foundation of Hunan Province (No. 2022JJ40799).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. D’Alterio, C.; Scala, S.; Sozzi, G.; Roz, L.; Bertolini, G. Paradoxical effects of chemotherapy on tumor relapse and metastasis promotion. Semin. Cancer Biol. 2020, 60, 351–361. [Google Scholar] [CrossRef] [PubMed]
  2. Garcia-Mayea, Y.; Mir, C.; Masson, F.; Paciucci, R.; ME, L.L. Insights into new mechanisms and models of cancer stem cell multidrug resistance. Semin. Cancer Biol. 2020, 60, 166–180. [Google Scholar] [CrossRef] [PubMed]
  3. Walcher, L.; Kistenmacher, A.K.; Suo, H.; Kitte, R.; Dluczek, S.; Strauss, A.; Blaudszun, A.R.; Yevsa, T.; Fricke, S.; Kossatz-Boehlert, U. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front. Immunol. 2020, 11, 1280. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, D.; Li, Y.; Ge, H.; Ghadban, T.; Reeh, M.; Gungor, C. The Extracellular Matrix: A Key Accomplice of Cancer Stem Cell Migration, Metastasis Formation, and Drug Resistance in PDAC. Cancers 2022, 14, 3998. [Google Scholar] [CrossRef]
  5. Voog, J.; Jones, D.L. Stem cells and the niche: A dynamic duo. Cell Stem Cell 2010, 6, 103–115. [Google Scholar] [CrossRef] [Green Version]
  6. Jin, J.; Bakker, A.D.; Wu, G.; Klein-Nulend, J.; Jaspers, R.T. Physicochemical Niche Conditions and Mechanosensing by Osteocytes and Myocytes. Curr. Osteoporos. Rep. 2019, 17, 235–249. [Google Scholar] [CrossRef] [Green Version]
  7. Morrison, S.J.; Kimble, J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 2006, 441, 1068–1074. [Google Scholar] [CrossRef]
  8. Li, X.; Wang, J. Mechanical tumor microenvironment and transduction: Cytoskeleton mediates cancer cell invasion and metastasis. Int. J. Biol. Sci. 2020, 16, 2014–2028. [Google Scholar] [CrossRef] [PubMed]
  9. Fife, C.M.; McCarroll, J.A.; Kavallaris, M. Movers and shakers: Cell cytoskeleton in cancer metastasis. Br. J. Pharmacol. 2014, 171, 5507–5523. [Google Scholar] [CrossRef] [Green Version]
  10. Strube, F.; Infanger, M.; Wehland, M.; Delvinioti, X.; Romswinkel, A.; Dietz, C.; Kraus, A. Alteration of Cytoskeleton Morphology and Gene Expression in Human Breast Cancer Cells under Simulated Microgravity. Cell J. 2020, 22, 106–114. [Google Scholar] [CrossRef]
  11. Samardzija, C.; Greening, D.W.; Escalona, R.; Chen, M.; Bilandzic, M.; Luwor, R.; Kannourakis, G.; Findlay, J.K.; Ahmed, N. Knockdown of stem cell regulator Oct4A in ovarian cancer reveals cellular reprogramming associated with key regulators of cytoskeleton-extracellular matrix remodelling. Sci. Rep. 2017, 7, 46312. [Google Scholar] [CrossRef] [PubMed]
  12. Kim, J.; Cheong, J.H. Role of Mitochondria-Cytoskeleton Interactions in the Regulation of Mitochondrial Structure and Function in Cancer Stem Cells. Cells 2020, 9, 1691. [Google Scholar] [CrossRef] [PubMed]
  13. Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef] [PubMed]
  14. Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef]
  15. So, J.Y.; Suh, N. Targeting cancer stem cells in solid tumors by vitamin D. J. Steroid Biochem. Mol. Biol. 2015, 148, 79–85. [Google Scholar] [CrossRef] [Green Version]
  16. Sulaiman, A.; McGarry, S.; Han, X.; Liu, S.; Wang, L. CSCs in Breast Cancer-One Size Does Not Fit All: Therapeutic Advances in Targeting Heterogeneous Epithelial and Mesenchymal CSCs. Cancers 2019, 11, 1128. [Google Scholar] [CrossRef] [Green Version]
  17. Shibata, M.; Hoque, M.O. Targeting Cancer Stem Cells: A Strategy for Effective Eradication of Cancer. Cancers 2019, 11, 732. [Google Scholar] [CrossRef] [Green Version]
  18. Matsui, W.; Huff, C.A.; Wang, Q.; Malehorn, M.T.; Barber, J.; Tanhehco, Y.; Smith, B.D.; Civin, C.I.; Jones, R.J. Characterization of clonogenic multiple myeloma cells. Blood 2004, 103, 2332–2336. [Google Scholar] [CrossRef] [Green Version]
  19. Bjerkvig, R.; Tysnes, B.B.; Aboody, K.S.; Najbauer, J.; Terzis, A.J. Opinion: The origin of the cancer stem cell: Current controversies and new insights. Nat. Rev. Cancer 2005, 5, 899–904. [Google Scholar] [CrossRef]
  20. Hill, R.P. Identifying cancer stem cells in solid tumors: Case not proven. Cancer Res. 2006, 66, 1891–1895. [Google Scholar] [CrossRef]
  21. Kanwar, S.S.; Yu, Y.; Nautiyal, J.; Patel, B.B.; Majumdar, A.P. The Wnt/beta-catenin pathway regulates growth and maintenance of colonospheres. Mol. Cancer 2010, 9, 212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wang, J.; Wakeman, T.P.; Lathia, J.D.; Hjelmeland, A.B.; Wang, X.F.; White, R.R.; Rich, J.N.; Sullenger, B.A. Notch promotes radioresistance of glioma stem cells. Stem Cells 2010, 28, 17–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Li, C.; Heidt, D.G.; Dalerba, P.; Burant, C.F.; Zhang, L.; Adsay, V.; Wicha, M.; Clarke, M.F.; Simeone, D.M. Identification of pancreatic cancer stem cells. Cancer Res. 2007, 67, 1030–1037. [Google Scholar] [CrossRef] [Green Version]
  24. Nguyen, L.V.; Vanner, R.; Dirks, P.; Eaves, C.J. Cancer stem cells: An evolving concept. Nat. Rev. Cancer 2012, 12, 133–143. [Google Scholar] [CrossRef] [PubMed]
  25. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef] [Green Version]
  26. Jin, X.; Jin, X.; Kim, H. Cancer stem cells and differentiation therapy. Tumour Biol. 2017, 39, 1010428317729933. [Google Scholar] [CrossRef] [Green Version]
  27. Talukdar, S.; Bhoopathi, P.; Emdad, L.; Das, S.; Sarkar, D.; Fisher, P.B. Dormancy and cancer stem cells: An enigma for cancer therapeutic targeting. Adv. Cancer Res. 2019, 141, 43–84. [Google Scholar] [CrossRef]
  28. Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [Green Version]
  29. Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 2019, 18, 157. [Google Scholar] [CrossRef] [Green Version]
  30. Lee, E.; Yang, J.; Ku, M.; Kim, N.H.; Park, Y.; Park, C.B.; Suh, J.S.; Park, E.S.; Yook, J.I.; Mills, G.B.; et al. Metabolic stress induces a Wnt-dependent cancer stem cell-like state transition. Cell Death Dis. 2015, 6, e1805. [Google Scholar] [CrossRef]
  31. Peiris-Pages, M.; Martinez-Outschoorn, U.E.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer stem cell metabolism. Breast Cancer Res. 2016, 18, 55. [Google Scholar] [CrossRef] [PubMed]
  32. Obre, E.; Rossignol, R. Emerging concepts in bioenergetics and cancer research: Metabolic flexibility, coupling, symbiosis, switch, oxidative tumors, metabolic remodeling, signaling and bioenergetic therapy. Int. J. Biochem. Cell Biol. 2015, 59, 167–181. [Google Scholar] [CrossRef] [PubMed]
  33. Kim, S.Y. Targeting cancer energy metabolism: A potential systemic cure for cancer. Arch. Pharm. Res. 2019, 42, 140–149. [Google Scholar] [CrossRef] [PubMed]
  34. Kroemer, G.; Pouyssegur, J. Tumor cell metabolism: Cancer’s Achilles’ heel. Cancer Cell 2008, 13, 472–482. [Google Scholar] [CrossRef]
  35. Araujo, E.P.; Carvalheira, J.B.; Velloso, L.A. Disruption of metabolic pathways--perspectives for the treatment of cancer. Curr. Cancer Drug Targets 2006, 6, 77–87. [Google Scholar] [CrossRef]
  36. Janiszewska, M.; Suva, M.L.; Riggi, N.; Houtkooper, R.H.; Auwerx, J.; Clement-Schatlo, V.; Radovanovic, I.; Rheinbay, E.; Provero, P.; Stamenkovic, I. Imp2 controls oxidative phosphorylation and is crucial for preserving glioblastoma cancer stem cells. Genes Dev. 2012, 26, 1926–1944. [Google Scholar] [CrossRef] [Green Version]
  37. Pasto, A.; Bellio, C.; Pilotto, G.; Ciminale, V.; Silic-Benussi, M.; Guzzo, G.; Rasola, A.; Frasson, C.; Nardo, G.; Zulato, E.; et al. Cancer stem cells from epithelial ovarian cancer patients privilege oxidative phosphorylation, and resist glucose deprivation. Oncotarget 2014, 5, 4305–4319. [Google Scholar] [CrossRef] [Green Version]
  38. Shen, Y.A.; Lin, C.H.; Chi, W.H.; Wang, C.Y.; Hsieh, Y.T.; Wei, Y.H.; Chen, Y.J. Resveratrol Impedes the Stemness, Epithelial-Mesenchymal Transition, and Metabolic Reprogramming of Cancer Stem Cells in Nasopharyngeal Carcinoma through p53 Activation. Evid. Based Complement. Alternat. Med. 2013, 2013, 590393. [Google Scholar] [CrossRef] [Green Version]
  39. Lagadinou, E.D.; Sach, A.; Callahan, K.; Rossi, R.M.; Neering, S.J.; Minhajuddin, M.; Ashton, J.M.; Pei, S.; Grose, V.; O’Dwyer, K.M.; et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell 2013, 12, 329–341. [Google Scholar] [CrossRef] [Green Version]
  40. Ye, X.Q.; Li, Q.; Wang, G.H.; Sun, F.F.; Huang, G.J.; Bian, X.W.; Yu, S.C.; Qian, G.S. Mitochondrial and energy metabolism-related properties as novel indicators of lung cancer stem cells. Int. J. Cancer 2011, 129, 820–831. [Google Scholar] [CrossRef]
  41. Farnie, G.; Sotgia, F.; Lisanti, M.P. High mitochondrial mass identifies a sub-population of stem-like cancer cells that are chemo-resistant. Oncotarget 2015, 6, 30472–30486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Chen, Y.; Li, D.; Wang, D.; Liu, X.; Yin, N.; Song, Y.; Lu, S.H.; Ju, Z.; Zhan, Q. Quiescence and attenuated DNA damage response promote survival of esophageal cancer stem cells. J. Cell. Biochem. 2012, 113, 3643–3652. [Google Scholar] [CrossRef] [PubMed]
  43. Kreso, A.; O’Brien, C.A.; van Galen, P.; Gan, O.I.; Notta, F.; Brown, A.M.; Ng, K.; Ma, J.; Wienholds, E.; Dunant, C.; et al. Variable clonal repopulation dynamics influence chemotherapy response in colorectal cancer. Science 2013, 339, 543–548. [Google Scholar] [CrossRef] [Green Version]
  44. Erin, N.; Grahovac, J.; Brozovic, A.; Efferth, T. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist. Updates 2020, 53, 100715. [Google Scholar] [CrossRef]
  45. Shibue, T.; Weinberg, R.A. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 611–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Nogales, E. Structural insight into microtubule function. Annu. Rev. Biophys. Biomol. Struct. 2001, 30, 397–420. [Google Scholar] [CrossRef]
  47. Heald, R.; Nogales, E. Microtubule dynamics. J. Cell Sci. 2002, 115, 3–4. [Google Scholar] [CrossRef]
  48. Popowicz, G.M.; Schleicher, M.; Noegel, A.A.; Holak, T.A. Filamins: Promiscuous organizers of the cytoskeleton. Trends Biochem. Sci. 2006, 31, 411–419. [Google Scholar] [CrossRef]
  49. Grintsevich, E.E.; Ahmed, G.; Ginosyan, A.A.; Wu, H.; Rich, S.K.; Reisler, E.; Terman, J.R. Profilin and Mical combine to impair F-actin assembly and promote disassembly and remodeling. Nat. Commun. 2021, 12, 5542. [Google Scholar] [CrossRef]
  50. Merino, F.; Pospich, S.; Raunser, S. Towards a structural understanding of the remodeling of the actin cytoskeleton. Semin. Cell Dev. Biol. 2020, 102, 51–64. [Google Scholar] [CrossRef]
  51. Ofer, N.; Abu Shah, E.; Keren, K. Differential mapping of the free barbed and pointed ends of actin filaments in cells. Cytoskeleton 2014, 71, 341–350. [Google Scholar] [CrossRef] [PubMed]
  52. Bisaria, A.; Hayer, A.; Garbett, D.; Cohen, D.; Meyer, T. Membrane-proximal F-actin restricts local membrane protrusions and directs cell migration. Science 2020, 368, 1205–1210. [Google Scholar] [CrossRef] [PubMed]
  53. Manor, U.; Bartholomew, S.; Golani, G.; Christenson, E.; Kozlov, M.; Higgs, H.; Spudich, J.; Lippincott-Schwartz, J. A mitochondria-anchored isoform of the actin-nucleating spire protein regulates mitochondrial division. Elife 2015, 4, e08828. [Google Scholar] [CrossRef] [PubMed]
  54. Moore, A.S.; Wong, Y.C.; Simpson, C.L.; Holzbaur, E.L. Dynamic actin cycling through mitochondrial subpopulations locally regulates the fission-fusion balance within mitochondrial networks. Nat. Commun. 2016, 7, 12886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Kruppa, A.J.; Kishi-Itakura, C.; Masters, T.A.; Rorbach, J.E.; Grice, G.L.; Kendrick-Jones, J.; Nathan, J.A.; Minczuk, M.; Buss, F. Myosin VI-Dependent Actin Cages Encapsulate Parkin-Positive Damaged Mitochondria. Dev. Cell 2018, 44, 484–499.e486. [Google Scholar] [CrossRef] [Green Version]
  56. Valencia, D.A.; Quinlan, M.E. Formins. Curr. Biol. 2021, 31, R517–R522. [Google Scholar] [CrossRef]
  57. Izdebska, M.; Zielinska, W.; Halas-Wisniewska, M.; Grzanka, A. Involvement of Actin and Actin-Binding Proteins in Carcinogenesis. Cells 2020, 9, 2245. [Google Scholar] [CrossRef]
  58. Izdebska, M.; Zielinska, W.; Grzanka, D.; Gagat, M. The Role of Actin Dynamics and Actin-Binding Proteins Expression in Epithelial-to-Mesenchymal Transition and Its Association with Cancer Progression and Evaluation of Possible Therapeutic Targets. Biomed. Res. Int. 2018, 2018, 4578373. [Google Scholar] [CrossRef] [Green Version]
  59. Namgoong, S.; Kim, N.H. Roles of actin binding proteins in mammalian oocyte maturation and beyond. Cell Cycle 2016, 15, 1830–1843. [Google Scholar] [CrossRef] [Green Version]
  60. Lee, C.W.; Vitriol, E.A.; Shim, S.; Wise, A.L.; Velayutham, R.P.; Zheng, J.Q. Dynamic localization of G-actin during membrane protrusion in neuronal motility. Curr. Biol. 2013, 23, 1046–1056. [Google Scholar] [CrossRef]
  61. Pinto-Costa, R.; Sousa, M.M. Profilin as a dual regulator of actin and microtubule dynamics. Cytoskeleton 2020, 77, 76–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Schmidt, E.J.; Funes, S.; McKeon, J.E.; Morgan, B.R.; Boopathy, S.; O’Connor, L.C.; Bilsel, O.; Massi, F.; Jegou, A.; Bosco, D.A. ALS-linked PFN1 variants exhibit loss and gain of functions in the context of formin-induced actin polymerization. Proc. Natl. Acad. Sci. USA 2021, 118, e2024605118. [Google Scholar] [CrossRef] [PubMed]
  63. Skruber, K.; Read, T.A.; Vitriol, E.A. Reconsidering an active role for G-actin in cytoskeletal regulation. J. Cell Sci. 2018, 131, jcs203760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Coumans, J.V.F.; Davey, R.J.; Moens, P.D.J. Cofilin and profilin: Partners in cancer aggressiveness. Biophys. Rev. 2018, 10, 1323–1335. [Google Scholar] [CrossRef]
  65. Vartiainen, M.K.; Sarkkinen, E.M.; Matilainen, T.; Salminen, M.; Lappalainen, P. Mammals have two twinfilin isoforms whose subcellular localizations and tissue distributions are differentially regulated. J. Biol. Chem. 2003, 278, 34347–34355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Poukkula, M.; Kremneva, E.; Serlachius, M.; Lappalainen, P. Actin-depolymerizing factor homology domain: A conserved fold performing diverse roles in cytoskeletal dynamics. Cytoskeleton 2011, 68, 471–490. [Google Scholar] [CrossRef]
  67. Becker, I.C.; Scheller, I.; Wackerbarth, L.M.; Beck, S.; Heib, T.; Aurbach, K.; Manukjan, G.; Gross, C.; Spindler, M.; Nagy, Z.; et al. Actin/microtubule crosstalk during platelet biogenesis in mice is critically regulated by Twinfilin1 and Cofilin1. Blood Adv. 2020, 4, 2124–2134. [Google Scholar] [CrossRef]
  68. Johnston, A.B.; Hilton, D.M.; McConnell, P.; Johnson, B.; Harris, M.T.; Simone, A.; Amarasinghe, G.K.; Cooper, J.A.; Goode, B.L. A novel mode of capping protein-regulation by twinfilin. Elife 2018, 7, e41313. [Google Scholar] [CrossRef]
  69. Kaishang, Z.; Xue, P.; Shaozhong, Z.; Yingying, F.; Yan, Z.; Chanjun, S.; Zhenzhen, L.; Xiangnan, L. Elevated expression of Twinfilin-1 is correlated with inferior prognosis of lung adenocarcinoma. Life Sci. 2018, 215, 159–169. [Google Scholar] [CrossRef]
  70. Husson, C.; Cantrelle, F.X.; Roblin, P.; Didry, D.; Le, K.H.; Perez, J.; Guittet, E.; Van Heijenoort, C.; Renault, L.; Carlier, M.F. Multifunctionality of the beta-thymosin/WH2 module: G-actin sequestration, actin filament growth, nucleation, and severing. Ann. N. Y. Acad. Sci. 2010, 1194, 44–52. [Google Scholar] [CrossRef]
  71. Bjorklund, G.; Dadar, M.; Aaseth, J.; Chirumbolo, S. Thymosin beta4: A Multi-Faceted Tissue Repair Stimulating Protein in Heart Injury. Curr. Med. Chem. 2020, 27, 6294–6305. [Google Scholar] [CrossRef] [PubMed]
  72. Zhao, K.N.; Masci, P.P.; Lavin, M.F. Disruption of spectrin-like cytoskeleton in differentiating keratinocytes by PKCdelta activation is associated with phosphorylated adducin. PLoS ONE 2011, 6, e28267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Oh, J.M.; Moon, E.Y. Actin-sequestering protein, thymosin beta-4, induces paclitaxel resistance through ROS/HIF-1alpha stabilization in HeLa human cervical tumor cells. Life Sci. 2010, 87, 286–293. [Google Scholar] [CrossRef] [PubMed]
  74. Hong, K.O.; Lee, J.I.; Hong, S.P.; Hong, S.D. Thymosin beta4 induces proliferation, invasion, and epithelial-to-mesenchymal transition of oral squamous cell carcinoma. Amino Acids 2016, 48, 117–127. [Google Scholar] [CrossRef] [Green Version]
  75. Lv, S.; Cai, H.; Xu, Y.; Dai, J.; Rong, X.; Zheng, L. Thymosinbeta 4 induces angiogenesis in critical limb ischemia mice via regulating Notch/NFkappaB pathway. Int. J. Mol. Med. 2020, 46, 1347–1358. [Google Scholar] [CrossRef]
  76. Kudryashova, E.; Heisler, D.B.; Williams, B.; Harker, A.J.; Shafer, K.; Quinlan, M.E.; Kovar, D.R.; Vavylonis, D.; Kudryashov, D.S. Actin Cross-Linking Toxin Is a Universal Inhibitor of Tandem-Organized and Oligomeric G-Actin Binding Proteins. Curr. Biol. 2018, 28, 1536–1547.e1539. [Google Scholar] [CrossRef] [Green Version]
  77. Jayo, A.; Malboubi, M.; Antoku, S.; Chang, W.; Ortiz-Zapater, E.; Groen, C.; Pfisterer, K.; Tootle, T.; Charras, G.; Gundersen, G.G.; et al. Fascin Regulates Nuclear Movement and Deformation in Migrating Cells. Dev. Cell 2016, 38, 371–383. [Google Scholar] [CrossRef] [Green Version]
  78. Gallop, J.L. Filopodia and their links with membrane traffic and cell adhesion. Semin. Cell Dev. Biol. 2020, 102, 81–89. [Google Scholar] [CrossRef]
  79. Lin, S.; Taylor, M.D.; Singh, P.K.; Yang, S. How does fascin promote cancer metastasis? FEBS J. 2021, 288, 1434–1446. [Google Scholar] [CrossRef]
  80. Cardama, G.A.; Gonzalez, N.; Maggio, J.; Menna, P.L.; Gomez, D.E. Rho GTPases as therapeutic targets in cancer (Review). Int. J. Oncol. 2017, 51, 1025–1034. [Google Scholar] [CrossRef]
  81. Parsons, M.; Adams, J.C. Rac regulates the interaction of fascin with protein kinase C in cell migration. J. Cell Sci. 2008, 121, 2805–2813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Jayo, A.; Parsons, M.; Adams, J.C. A novel Rho-dependent pathway that drives interaction of fascin-1 with p-Lin-11/Isl-1/Mec-3 kinase (LIMK) 1/2 to promote fascin-1/actin binding and filopodia stability. BMC Biol. 2012, 10, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Gonzalez-Morales, N.; Holenka, T.K.; Schock, F. Filamin actin-binding and titin-binding fulfill distinct functions in Z-disc cohesion. PLoS Genet. 2017, 13, e1006880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Wang, X.; Zhu, H.; Lu, Y.; Wang, Z.; Kennedy, D. The elastic properties and deformation mechanisms of actin filament networks crosslinked by filamins. J. Mech. Behav. Biomed. Mater. 2020, 112, 104075. [Google Scholar] [CrossRef] [PubMed]
  85. Lad, Y.; Kiema, T.; Jiang, P.; Pentikainen, O.T.; Coles, C.H.; Campbell, I.D.; Calderwood, D.A.; Ylanne, J. Structure of three tandem filamin domains reveals auto-inhibition of ligand binding. EMBO J. 2007, 26, 3993–4004. [Google Scholar] [CrossRef] [Green Version]
  86. Svitkina, T. The Actin Cytoskeleton and Actin-Based Motility. Cold Spring Harb. Perspect. Biol. 2018, 10, a018267. [Google Scholar] [CrossRef] [Green Version]
  87. Iwamoto, D.V.; Huehn, A.; Simon, B.; Huet-Calderwood, C.; Baldassarre, M.; Sindelar, C.V.; Calderwood, D.A. Structural basis of the filamin A actin-binding domain interaction with F-actin. Nat. Struct. Mol. Biol. 2018, 25, 918–927. [Google Scholar] [CrossRef] [Green Version]
  88. Tirupula, K.C.; Ithychanda, S.S.; Mohan, M.L.; Naga Prasad, S.V.; Qin, J.; Karnik, S.S. G protein-coupled receptors directly bind filamin A with high affinity and promote filamin phosphorylation. Biochemistry 2015, 54, 6673–6683. [Google Scholar] [CrossRef]
  89. Liem, R.K. Cytoskeletal Integrators: The Spectrin Superfamily. Cold Spring Harb. Perspect. Biol. 2016, 8, a018259. [Google Scholar] [CrossRef] [Green Version]
  90. Zhang, R.; Zhang, C.; Zhao, Q.; Li, D. Spectrin: Structure, function and disease. Sci. China Life Sci. 2013, 56, 1076–1085. [Google Scholar] [CrossRef]
  91. Unudurthi, S.D.; Greer-Short, A.; Patel, N.; Nassal, D.; Hund, T.J. Spectrin-based pathways underlying electrical and mechanical dysfunction in cardiac disease. Expert Rev. Cardiovasc. Ther. 2018, 16, 59–65. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, Y.; Yago, T.; Zhang, N.; Abdisalaam, S.; Alexandrakis, G.; Rodgers, W.; McEver, R.P. Cytoskeletal regulation of CD44 membrane organization and interactions with E-selectin. J. Biol. Chem. 2014, 289, 35159–35171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Aseervatham, J. Cytoskeletal Remodeling in Cancer. Biology 2020, 9, 385. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, Z.; Song, Y.; Tu, W.; He, X.; Lin, J.; Liu, F. beta-2 spectrin is involved in hepatocyte proliferation through the interaction of TGFbeta/Smad and PI3K/AKT signalling. Liver Int. 2012, 32, 1103–1111. [Google Scholar] [CrossRef]
  95. Wu, X.T.; Sun, L.W.; Yang, X.; Ding, D.; Han, D.; Fan, Y.B. The potential role of spectrin network in the mechanotransduction of MLO-Y4 osteocytes. Sci. Rep. 2017, 7, 40940. [Google Scholar] [CrossRef] [Green Version]
  96. Murphy, A.C.; Young, P.W. The actinin family of actin cross-linking proteins—A genetic perspective. Cell Biosci. 2015, 5, 49. [Google Scholar] [CrossRef] [Green Version]
  97. Thomas, D.G.; Robinson, D.N. The fifth sense: Mechanosensory regulation of alpha-actinin-4 and its relevance for cancer metastasis. Semin. Cell Dev. Biol. 2017, 71, 68–74. [Google Scholar] [CrossRef]
  98. Nagano, M.; Hoshino, D.; Koshikawa, N.; Akizawa, T.; Seiki, M. Turnover of focal adhesions and cancer cell migration. Int. J. Cell Biol. 2012, 2012, 310616. [Google Scholar] [CrossRef] [Green Version]
  99. Castellano, E.; Downward, J. RAS Interaction with PI3K: More Than Just Another Effector Pathway. Genes Cancer 2011, 2, 261–274. [Google Scholar] [CrossRef] [Green Version]
  100. Deming, P.B.; Campbell, S.L.; Stone, J.B.; Rivard, R.L.; Mercier, A.L.; Howe, A.K. Anchoring of protein kinase A by ERM (ezrin-radixin-moesin) proteins is required for proper netrin signaling through DCC (deleted in colorectal cancer). J. Biol. Chem. 2015, 290, 5783–5796. [Google Scholar] [CrossRef]
  101. Gao, S.; Dai, Y.; Yin, M.; Ye, J.; Li, G.; Yu, J. Potential transcriptional regulatory regions exist upstream of the human ezrin gene promoter in esophageal carcinoma cells. Acta Biochim. Biophys. Sin. 2011, 43, 455–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Biri-Kovacs, B.; Kiss, B.; Vadaszi, H.; Gogl, G.; Palfy, G.; Torok, G.; Homolya, L.; Bodor, A.; Nyitray, L. Ezrin interacts with S100A4 via both its N- and C-terminal domains. PLoS ONE 2017, 12, e0177489. [Google Scholar] [CrossRef] [Green Version]
  103. Lema, B.E.; Patricio, G.M.; Kreimann, E.L. Nuclear expression of NHERF1/EBP50 in Clear Cell Renal Cell Carcinoma. Acta Histochem. 2021, 123, 151717. [Google Scholar] [CrossRef] [PubMed]
  104. Stevenson, R.P.; Veltman, D.; Machesky, L.M. Actin-bundling proteins in cancer progression at a glance. J. Cell Sci. 2012, 125, 1073–1079. [Google Scholar] [CrossRef] [Green Version]
  105. Shi, K.; Yang, L.; Du, X.; Guo, D.; Xue, L. Molecular chaperone Hsp90 protects KCBP from degradation by proteasome in Dunaliella salina cells. Folia Microbiol. 2021, 66, 949–957. [Google Scholar] [CrossRef] [PubMed]
  106. Kamioka, H.; Tomono, T.; Fujita, A.; Onozato, R.; Iijima, M.; Tsuchida, S.; Arai, T.; Fujita, Y.; Zhang, X.; Yano, K.; et al. Moesin-Mediated P-Glycoprotein Activation During Snail-Induced Epithelial-Mesenchymal Transition in Lung Cancer Cells. J. Pharm. Sci. 2020, 109, 2302–2308. [Google Scholar] [CrossRef]
  107. Rahimi, N.; Ho, R.X.Y.; Chandler, K.B.; De La Cena, K.O.C.; Amraei, R.; Mitchel, A.J.; Engblom, N.; Costello, C.E. The cell adhesion molecule TMIGD1 binds to moesin and regulates tubulin acetylation and cell migration. J. Biomed. Sci. 2021, 28, 61. [Google Scholar] [CrossRef]
  108. Karvar, S.; Ansa-Addo, E.A.; Suda, J.; Singh, S.; Zhu, L.; Li, Z.; Rockey, D.C. Moesin, an Ezrin/Radixin/Moesin Family Member, Regulates Hepatic Fibrosis. Hepatology 2020, 72, 1073–1084. [Google Scholar] [CrossRef]
  109. Jiang, Q.H.; Wang, A.X.; Chen, Y. Radixin enhances colon cancer cell invasion by increasing MMP-7 production via Rac1-ERK pathway. Sci. World J. 2014, 2014, 340271. [Google Scholar] [CrossRef] [Green Version]
  110. Hoeflich, K.P.; Ikura, M. Radixin: Cytoskeletal adopter and signaling protein. Int. J. Biochem. Cell Biol. 2004, 36, 2131–2136. [Google Scholar] [CrossRef]
  111. Michie, K.A.; Bermeister, A.; Robertson, N.O.; Goodchild, S.C.; Curmi, P.M.G. Two Sides of the Coin: Ezrin/Radixin/Moesin and Merlin Control Membrane Structure and Contact Inhibition. Int. J. Mol. Sci. 2019, 20, 1996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Chinthalapudi, K.; Mandati, V.; Zheng, J.; Sharff, A.J.; Bricogne, G.; Griffin, P.R.; Kissil, J.; Izard, T. Lipid binding promotes the open conformation and tumor-suppressive activity of neurofibromin 2. Nat. Commun. 2018, 9, 1338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhang, Y.; Luo, X.; Lin, J.; Fu, S.; Feng, P.; Su, H.; He, X.; Liang, X.; Liu, K.; Deng, W. Gelsolin Promotes Cancer Progression by Regulating Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma and Correlates with a Poor Prognosis. J. Oncol. 2020, 2020, 1980368. [Google Scholar] [CrossRef] [PubMed]
  114. Nag, S.; Larsson, M.; Robinson, R.C.; Burtnick, L.D. Gelsolin: The tail of a molecular gymnast. Cytoskeleton 2013, 70, 360–384. [Google Scholar] [CrossRef] [PubMed]
  115. Morley, S.C.; Sung, J.; Sun, G.P.; Martelli, M.P.; Bunnell, S.C.; Bierer, B.E. Gelsolin overexpression alters actin dynamics and tyrosine phosphorylation of lipid raft-associated proteins in Jurkat T cells. Mol. Immunol. 2007, 44, 2469–2480. [Google Scholar] [CrossRef] [Green Version]
  116. Narita, A. ADF/cofilin regulation from a structural viewpoint. J. Muscle Res. Cell Motil. 2020, 41, 141–151. [Google Scholar] [CrossRef]
  117. Ostrowska, Z.; Moraczewska, J. Cofilin—A protein controlling dynamics of actin filaments. Postepy Hig. Med. Dosw. (Online) 2017, 71, 339–351. [Google Scholar] [CrossRef]
  118. Hamill, S.; Lou, H.J.; Turk, B.E.; Boggon, T.J. Structural Basis for Noncanonical Substrate Recognition of Cofilin/ADF Proteins by LIM Kinases. Mol. Cell 2016, 62, 397–408. [Google Scholar] [CrossRef] [Green Version]
  119. Kaushik, V.; Brunnert, D.; Hanschmann, E.M.; Sharma, P.K.; Anand, B.G.; Kar, K.; Kateriya, S.; Goyal, P. The intrinsic amyloidogenic propensity of cofilin-1 is aggravated by Cys-80 oxidation: A possible link with neurodegenerative diseases. Biochem. Biophys. Res. Commun. 2021, 569, 187–192. [Google Scholar] [CrossRef]
  120. Ubelmann, F.; Chamaillard, M.; El-Marjou, F.; Simon, A.; Netter, J.; Vignjevic, D.; Nichols, B.L.; Quezada-Calvillo, R.; Grandjean, T.; Louvard, D.; et al. Enterocyte loss of polarity and gut wound healing rely upon the F-actin-severing function of villin. Proc. Natl. Acad. Sci. USA 2013, 110, E1380–E1389. [Google Scholar] [CrossRef]
  121. Hampton, C.M.; Liu, J.; Taylor, D.W.; DeRosier, D.J.; Taylor, K.A. The 3D structure of villin as an unusual F-Actin crosslinker. Structure 2008, 16, 1882–1891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Ghoshdastider, U.; Popp, D.; Burtnick, L.D.; Robinson, R.C. The expanding superfamily of gelsolin homology domain proteins. Cytoskeleton 2013, 70, 775–795. [Google Scholar] [CrossRef] [PubMed]
  123. Chandra, S.; Kumar, M.; Sharma, N.R.; Sarkar, D.P. Site-specific phosphorylation of villin remodels the actin cytoskeleton to regulate Sendai viral glycoprotein-mediated membrane fusion. FEBS Lett. 2019, 593, 1927–1943. [Google Scholar] [CrossRef] [PubMed]
  124. Kumar, N.; Zhao, P.; Tomar, A.; Galea, C.A.; Khurana, S. Association of villin with phosphatidylinositol 4,5-bisphosphate regulates the actin cytoskeleton. J. Biol. Chem. 2004, 279, 3096–3110. [Google Scholar] [CrossRef] [Green Version]
  125. Fowler, V.M.; Dominguez, R. Tropomodulins and Leiomodins: Actin Pointed End Caps and Nucleators in Muscles. Biophys. J. 2017, 112, 1742–1760. [Google Scholar] [CrossRef] [Green Version]
  126. Ghosh, A.; Fowler, V.M. Tropomodulins. Curr. Biol. 2021, 31, R501–R503. [Google Scholar] [CrossRef]
  127. Ostrowska, Z.; Robaszkiewicz, K.; Moraczewska, J. Regulation of actin filament turnover by cofilin-1 and cytoplasmic tropomyosin isoforms. Biochim. Biophys. Acta Proteins Proteom. 2017, 1865, 88–98. [Google Scholar] [CrossRef]
  128. Gateva, G.; Tojkander, S.; Koho, S.; Carpen, O.; Lappalainen, P. Palladin promotes assembly of non-contractile dorsal stress fibers through VASP recruitment. J. Cell Sci. 2014, 127, 1887–1898. [Google Scholar] [CrossRef] [Green Version]
  129. Salazar, M.A.; Kwiatkowski, A.V.; Pellegrini, L.; Cestra, G.; Butler, M.H.; Rossman, K.L.; Serna, D.M.; Sondek, J.; Gertler, F.B.; De Camilli, P. Tuba, a novel protein containing bin/amphiphysin/Rvs and Dbl homology domains, links dynamin to regulation of the actin cytoskeleton. J. Biol. Chem. 2003, 278, 49031–49043. [Google Scholar] [CrossRef] [Green Version]
  130. Bear, J.E.; Gertler, F.B. Ena/VASP: Towards resolving a pointed controversy at the barbed end. J. Cell Sci. 2009, 122, 1947–1953. [Google Scholar] [CrossRef]
  131. Zhang, Y.T.; Xu, L.H.; Lu, Q.; Liu, K.P.; Liu, P.Y.; Ji, F.; Liu, X.M.; Ouyang, D.Y.; He, X.H. VASP activation via the Galpha13/RhoA/PKA pathway mediates cucurbitacin-B-induced actin aggregation and cofilin-actin rod formation. PLoS ONE 2014, 9, e93547. [Google Scholar] [CrossRef]
  132. Goodson, H.V.; Jonasson, E.M. Microtubules and Microtubule-Associated Proteins. Cold Spring Harb. Perspect. Biol. 2018, 10, a022608. [Google Scholar] [CrossRef] [PubMed]
  133. Janke, C.; Magiera, M.M. The tubulin code and its role in controlling microtubule properties and functions. Nat. Rev. Mol. Cell Biol. 2020, 21, 307–326. [Google Scholar] [CrossRef]
  134. Chaudhary, N.; Nagaraj, R. Tau fibrillogenesis. Subcell. Biochem. 2012, 65, 75–90. [Google Scholar] [CrossRef]
  135. Breuzard, G.; Pagano, A.; Bastonero, S.; Malesinski, S.; Parat, F.; Barbier, P.; Peyrot, V.; Kovacic, H. Tau regulates the microtubule-dependent migration of glioblastoma cells via the Rho-ROCK signaling pathway. J. Cell Sci. 2019, 132, jcs222851. [Google Scholar] [CrossRef] [Green Version]
  136. Teng, J.; Takei, Y.; Harada, A.; Nakata, T.; Chen, J.; Hirokawa, N. Synergistic effects of MAP2 and MAP1B knockout in neuronal migration, dendritic outgrowth, and microtubule organization. J. Cell Biol. 2001, 155, 65–76. [Google Scholar] [CrossRef] [Green Version]
  137. Zehr, E.; Szyk, A.; Piszczek, G.; Szczesna, E.; Zuo, X.; Roll-Mecak, A. Katanin spiral and ring structures shed light on power stroke for microtubule severing. Nat. Struct. Mol. Biol. 2017, 24, 717–725. [Google Scholar] [CrossRef] [PubMed]
  138. Nehlig, A.; Seiler, C.; Steblyanko, Y.; Dingli, F.; Arras, G.; Loew, D.; Welburn, J.; Prigent, C.; Barisic, M.; Nahmias, C. Reciprocal regulation of Aurora kinase A and ATIP3 in the control of metaphase spindle length. Cell Mol. Life Sci. 2021, 78, 1765–1779. [Google Scholar] [CrossRef]
  139. Lu, W.; Gelfand, V.I. Moonlighting Motors: Kinesin, Dynein, and Cell Polarity. Trends Cell Biol. 2017, 27, 505–514. [Google Scholar] [CrossRef] [Green Version]
  140. Gigant, B.; Wang, W.; Dreier, B.; Jiang, Q.; Pecqueur, L.; Pluckthun, A.; Wang, C.; Knossow, M. Structure of a kinesin-tubulin complex and implications for kinesin motility. Nat. Struct. Mol. Biol. 2013, 20, 1001–1007. [Google Scholar] [CrossRef]
  141. Arora, K.; Talje, L.; Asenjo, A.B.; Andersen, P.; Atchia, K.; Joshi, M.; Sosa, H.; Allingham, J.S.; Kwok, B.H. KIF14 binds tightly to microtubules and adopts a rigor-like conformation. J. Mol. Biol. 2014, 426, 2997–3015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Roberts, A.J.; Kon, T.; Knight, P.J.; Sutoh, K.; Burgess, S.A. Functions and mechanics of dynein motor proteins. Nat. Rev. Mol. Cell Biol. 2013, 14, 713–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Wattanathamsan, O.; Pongrakhananon, V. Emerging role of microtubule-associated proteins on cancer metastasis. Front. Pharmacol. 2022, 13, 935493. [Google Scholar] [CrossRef] [PubMed]
  144. Zeitz, M.; Kierfeld, J. Feedback mechanism for microtubule length regulation by stathmin gradients. Biophys. J. 2014, 107, 2860–2871. [Google Scholar] [CrossRef] [Green Version]
  145. Gupta, K.K.; Li, C.; Duan, A.; Alberico, E.O.; Kim, O.V.; Alber, M.S.; Goodson, H.V. Mechanism for the catastrophe-promoting activity of the microtubule destabilizer Op18/stathmin. Proc. Natl. Acad. Sci. USA 2013, 110, 20449–20454. [Google Scholar] [CrossRef] [Green Version]
  146. Matov, A.; Applegate, K.; Kumar, P.; Thoma, C.; Krek, W.; Danuser, G.; Wittmann, T. Analysis of microtubule dynamic instability using a plus-end growth marker. NaMethods 2010, 7, 761–768. [Google Scholar] [CrossRef] [Green Version]
  147. Nehlig, A.; Molina, A.; Rodrigues-Ferreira, S.; Honore, S.; Nahmias, C. Regulation of end-binding protein EB1 in the control of microtubule dynamics. Cell Mol. Life Sci. 2017, 74, 2381–2393. [Google Scholar] [CrossRef] [Green Version]
  148. Lindeboom, J.J.; Nakamura, M.; Saltini, M.; Hibbel, A.; Walia, A.; Ketelaar, T.; Emons, A.M.C.; Sedbrook, J.C.; Kirik, V.; Mulder, B.M.; et al. CLASP stabilization of plus ends created by severing promotes microtubule creation and reorientation. J. Cell Biol. 2019, 218, 190–205. [Google Scholar] [CrossRef] [Green Version]
  149. Akhmanova, A.; Hoogenraad, C.C. Microtubule minus-end-targeting proteins. Curr. Biol. 2015, 25, R162–R171. [Google Scholar] [CrossRef] [Green Version]
  150. Atherton, J.; Jiang, K.; Stangier, M.M.; Luo, Y.; Hua, S.; Houben, K.; van Hooff, J.J.E.; Joseph, A.P.; Scarabelli, G.; Grant, B.J.; et al. A structural model for microtubule minus-end recognition and protection by CAMSAP proteins. Nat. Struct. Mol. Biol. 2017, 24, 931–943. [Google Scholar] [CrossRef]
  151. Guhathakurta, P.; Prochniewicz, E.; Thomas, D.D. Actin-Myosin Interaction: Structure, Function and Drug Discovery. Int. J. Mol. Sci. 2018, 19, 2628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Walker, B.C.; Walczak, C.E.; Cochran, J.C. Switch-1 instability at the active site decouples ATP hydrolysis from force generation in myosin II. Cytoskeleton 2021, 78, 3–13. [Google Scholar] [CrossRef] [PubMed]
  153. Cross, J.A.; Dodding, M.P. Motor-cargo adaptors at the organelle-cytoskeleton interface. Curr. Opin. Cell Biol. 2019, 59, 16–23. [Google Scholar] [CrossRef] [PubMed]
  154. Herrmann, H.; Aebi, U. Intermediate Filaments: Structure and Assembly. Cold Spring Harb. Perspect. Biol. 2016, 8, a018242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Chung, B.M.; Rotty, J.D.; Coulombe, P.A. Networking galore: Intermediate filaments and cell migration. Curr. Opin. Cell Biol. 2013, 25, 600–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Etienne-Manneville, S. Cytoplasmic Intermediate Filaments in Cell Biology. Annu. Rev. Cell Dev. Biol. 2018, 34, 1–28. [Google Scholar] [CrossRef]
  157. Hyder, C.L.; Pallari, H.M.; Kochin, V.; Eriksson, J.E. Providing cellular signposts—Post-translational modifications of intermediate filaments. FEBS Lett. 2008, 582, 2140–2148. [Google Scholar] [CrossRef] [Green Version]
  158. Leduc, C.; Etienne-Manneville, S. Intermediate filaments in cell migration and invasion: The unusual suspects. Curr. Opin. Cell Biol. 2015, 32, 102–112. [Google Scholar] [CrossRef]
  159. de Pereda, J.M.; Lillo, M.P.; Sonnenberg, A. Structural basis of the interaction between integrin alpha6beta4 and plectin at the hemidesmosomes. EMBO J. 2009, 28, 1180–1190. [Google Scholar] [CrossRef] [Green Version]
  160. Satelli, A.; Li, S. Vimentin in cancer and its potential as a molecular target for cancer therapy. Cell Mol. Life Sci. 2011, 68, 3033–3046. [Google Scholar] [CrossRef]
  161. Ambriz, X.; de Lanerolle, P.; Ambrosio, J.R. The Mechanobiology of the Actin Cytoskeleton in Stem Cells during Differentiation and Interaction with Biomaterials. Stem Cells Int. 2018, 2018, 2891957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Low, B.C.; Pan, C.Q.; Shivashankar, G.V.; Bershadsky, A.; Sudol, M.; Sheetz, M. YAP/TAZ as mechanosensors and mechanotransducers in regulating organ size and tumor growth. FEBS Lett. 2014, 588, 2663–2670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Yang, Y.M.; Chang, J.W. Current status and issues in cancer stem cell study. Cancer Investig. 2008, 26, 741–755. [Google Scholar] [CrossRef] [PubMed]
  164. Jabbari, E.; Sarvestani, S.K.; Daneshian, L.; Moeinzadeh, S. Optimum 3D Matrix Stiffness for Maintenance of Cancer Stem Cells Is Dependent on Tissue Origin of Cancer Cells. PLoS ONE 2015, 10, e0132377. [Google Scholar] [CrossRef] [Green Version]
  165. Bourguignon, L.Y.; Shiina, M.; Li, J.J. Hyaluronan-CD44 interaction promotes oncogenic signaling, microRNA functions, chemoresistance, and radiation resistance in cancer stem cells leading to tumor progression. Adv. Cancer Res. 2014, 123, 255–275. [Google Scholar] [CrossRef] [Green Version]
  166. Witke, W. The role of profilin complexes in cell motility and other cellular processes. Trends Cell Biol. 2004, 14, 461–469. [Google Scholar] [CrossRef]
  167. Kim, M.J.; Lee, Y.S.; Han, G.Y.; Lee, H.N.; Ahn, C.; Kim, C.W. Profilin 2 promotes migration, invasion, and stemness of HT29 human colorectal cancer stem cells. Biosci. Biotechnol. Biochem. 2015, 79, 1438–1446. [Google Scholar] [CrossRef] [Green Version]
  168. Pouremamali, F.; Vahedian, V.; Hassani, N.; Mirzaei, S.; Pouremamali, A.; Kazemzadeh, H.; Faridvand, Y.; Jafari-Gharabaghlou, D.; Nouri, M.; Maroufi, N.F. The role of SOX family in cancer stem cell maintenance: With a focus on SOX2. Pathol. Res. Pract. 2022, 231, 153783. [Google Scholar] [CrossRef]
  169. Jiang, C.; Ding, Z.; Joy, M.; Chakraborty, S.; Kim, S.H.; Bottcher, R.; Condeelis, J.; Singh, S.; Roy, P. A balanced level of profilin-1 promotes stemness and tumor-initiating potential of breast cancer cells. Cell Cycle 2017, 16, 2366–2373. [Google Scholar] [CrossRef] [Green Version]
  170. Kobayashi, T.; Okada, F.; Fujii, N.; Tomita, N.; Ito, S.; Tazawa, H.; Aoyama, T.; Choi, S.K.; Shibata, T.; Fujita, H.; et al. Thymosin-beta4 regulates motility and metastasis of malignant mouse fibrosarcoma cells. Am. J. Pathol. 2002, 160, 869–882. [Google Scholar] [CrossRef]
  171. Nemolato, S.; Restivo, A.; Cabras, T.; Coni, P.; Zorcolo, L.; Orru, G.; Fanari, M.; Cau, F.; Gerosa, C.; Fanni, D.; et al. Thymosin beta 4 in colorectal cancer is localized predominantly at the invasion front in tumor cells undergoing epithelial mesenchymal transition. Cancer Biol. Ther. 2012, 13, 191–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Ricci-Vitiani, L.; Mollinari, C.; di Martino, S.; Biffoni, M.; Pilozzi, E.; Pagliuca, A.; de Stefano, M.C.; Circo, R.; Merlo, D.; De Maria, R.; et al. Thymosin beta4 targeting impairs tumorigenic activity of colon cancer stem cells. FASEB J. 2010, 24, 4291–4301. [Google Scholar] [CrossRef] [PubMed]
  173. Ji, Y.I.; Lee, B.Y.; Kang, Y.J.; Jo, J.O.; Lee, S.H.; Kim, H.Y.; Kim, Y.O.; Lee, C.; Koh, S.B.; Kim, A.; et al. Expression patterns of Thymosin beta4 and cancer stem cell marker CD133 in ovarian cancers. Pathol. Oncol. Res. 2013, 19, 237–245. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, Y.; Feurino, L.W.; Zhai, Q.; Wang, H.; Fisher, W.E.; Chen, C.; Yao, Q.; Li, M. Thymosin Beta 4 is overexpressed in human pancreatic cancer cells and stimulates proinflammatory cytokine secretion and JNK activation. Cancer Biol. Ther. 2008, 7, 419–423. [Google Scholar] [CrossRef] [Green Version]
  175. Lee, S.I.; Kim, D.S.; Lee, H.J.; Cha, H.J.; Kim, E.C. The role of thymosin beta 4 on odontogenic differentiation in human dental pulp cells. PLoS ONE 2013, 8, e61960. [Google Scholar] [CrossRef] [Green Version]
  176. Bragdon, B.; Moseychuk, O.; Saldanha, S.; King, D.; Julian, J.; Nohe, A. Bone morphogenetic proteins: A critical review. Cell Signal 2011, 23, 609–620. [Google Scholar] [CrossRef]
  177. Tang, M.C.; Chan, L.C.; Yeh, Y.C.; Chen, C.Y.; Chou, T.Y.; Wang, W.S.; Su, Y. Thymosin beta 4 induces colon cancer cell migration and clinical metastasis via enhancing ILK/IQGAP1/Rac1 signal transduction pathway. Cancer Lett. 2011, 308, 162–171. [Google Scholar] [CrossRef]
  178. Samaeekia, R.; Adorno-Cruz, V.; Bockhorn, J.; Chang, Y.F.; Huang, S.; Prat, A.; Ha, N.; Kibria, G.; Huo, D.; Zheng, H.; et al. miR-206 Inhibits Stemness and Metastasis of Breast Cancer by Targeting MKL1/IL11 Pathway. Clin. Cancer Res. 2017, 23, 1091–1103. [Google Scholar] [CrossRef] [Green Version]
  179. Meacham, C.E.; Ho, E.E.; Dubrovsky, E.; Gertler, F.B.; Hemann, M.T. In vivo RNAi screening identifies regulators of actin dynamics as key determinants of lymphoma progression. Nat. Genet. 2009, 41, 1133–1137. [Google Scholar] [CrossRef] [Green Version]
  180. Barnawi, R.; Al-Khaldi, S.; Colak, D.; Tulbah, A.; Al-Tweigeri, T.; Fallatah, M.; Monies, D.; Ghebeh, H.; Al-Alwan, M. beta1 Integrin is essential for fascin-mediated breast cancer stem cell function and disease progression. Int. J. Cancer 2019, 145, 830–841. [Google Scholar] [CrossRef]
  181. Barnawi, R.; Al-Khaldi, S.; Bakheet, T.; Fallatah, M.; Alaiya, A.; Ghebeh, H.; Al-Alwan, M. Fascin Activates beta-Catenin Signaling and Promotes Breast Cancer Stem Cell Function Mainly Through Focal Adhesion Kinase (FAK): Relation with Disease Progression. Front. Oncol. 2020, 10, 440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Zhao, W.; Gao, J.; Wu, J.; Liu, Q.H.; Wang, Z.G.; Li, H.L.; Xing, L.H. Expression of Fascin-1 on human lung cancer and paracarcinoma tissue and its relation to clinicopathological characteristics in patients with lung cancer. Onco Targets Ther. 2015, 8, 2571–2576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. McGuire, S.; Kara, B.; Hart, P.C.; Montag, A.; Wroblewski, K.; Fazal, S.; Huang, X.Y.; Lengyel, E.; Kenny, H.A. Inhibition of fascin in cancer and stromal cells blocks ovarian cancer metastasis. Gynecol. Oncol. 2019, 153, 405–415. [Google Scholar] [CrossRef] [PubMed]
  184. Leung, R.; Wang, Y.; Cuddy, K.; Sun, C.; Magalhaes, J.; Grynpas, M.; Glogauer, M. Filamin A regulates monocyte migration through Rho small GTPases during osteoclastogenesis. J. Bone Miner Res. 2010, 25, 1077–1091. [Google Scholar] [CrossRef]
  185. Yue, J.; Lu, H.; Liu, J.; Berwick, M.; Shen, Z. Filamin-A as a marker and target for DNA damage based cancer therapy. DNA Repair 2012, 11, 192–200. [Google Scholar] [CrossRef] [Green Version]
  186. Savoy, R.M.; Ghosh, P.M. The dual role of filamin A in cancer: Can’t live with (too much of) it, can’t live without it. Endocr. Relat. Cancer 2013, 20, R341–R356. [Google Scholar] [CrossRef] [Green Version]
  187. Bourguignon, L.Y.W.; Earle, C.; Shiina, M. Activation of Matrix Hyaluronan-Mediated CD44 Signaling, Epigenetic Regulation and Chemoresistance in Head and Neck Cancer Stem Cells. Int. J. Mol. Sci. 2017, 18, 1849. [Google Scholar] [CrossRef] [Green Version]
  188. Zhang, R.; Liu, C.; Niu, Y.; Jing, Y.; Zhang, H.; Wang, J.; Yang, J.; Zen, K.; Zhang, J.; Zhang, C.Y.; et al. MicroRNA-128-3p regulates mitomycin C-induced DNA damage response in lung cancer cells through repressing SPTAN1. Oncotarget 2017, 8, 58098–58107. [Google Scholar] [CrossRef] [Green Version]
  189. Schrecker, C.; Behrens, S.; Schonherr, R.; Ackermann, A.; Pauli, D.; Plotz, G.; Zeuzem, S.; Brieger, A. SPTAN1 Expression Predicts Treatment and Survival Outcomes in Colorectal Cancer. Cancers 2021, 13, 3638. [Google Scholar] [CrossRef]
  190. Ackermann, A.; Schrecker, C.; Bon, D.; Friedrichs, N.; Bankov, K.; Wild, P.; Plotz, G.; Zeuzem, S.; Herrmann, E.; Hansmann, M.L.; et al. Downregulation of SPTAN1 is related to MLH1 deficiency and metastasis in colorectal cancer. PLoS ONE 2019, 14, e0213411. [Google Scholar] [CrossRef]
  191. Chen, Y.; Meng, L.; Shang, H.; Dou, Q.; Lu, Z.; Liu, L.; Wang, Z.; He, X.; Song, Y. beta2 spectrin-mediated differentiation repressed the properties of liver cancer stem cells through beta-catenin. Cell Death Dis. 2018, 9, 424. [Google Scholar] [CrossRef] [PubMed]
  192. Jung, J.; Kim, S.; An, H.T.; Ko, J. alpha-Actinin-4 regulates cancer stem cell properties and chemoresistance in cervical cancer. Carcinogenesis 2020, 41, 940–949. [Google Scholar] [CrossRef] [PubMed]
  193. Wang, N.; Wang, Q.; Tang, H.; Zhang, F.; Zheng, Y.; Wang, S.; Zhang, J.; Wang, Z.; Xie, X. Direct inhibition of ACTN4 by ellagic acid limits breast cancer metastasis via regulation of beta-catenin stabilization in cancer stem cells. J. Exp. Clin. Cancer Res. 2017, 36, 172. [Google Scholar] [CrossRef] [Green Version]
  194. Kikuchi, S.; Honda, K.; Tsuda, H.; Hiraoka, N.; Imoto, I.; Kosuge, T.; Umaki, T.; Onozato, K.; Shitashige, M.; Yamaguchi, U.; et al. Expression and gene amplification of actinin-4 in invasive ductal carcinoma of the pancreas. Clin. Cancer Res. 2008, 14, 5348–5356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Ren, L.; Hong, S.H.; Chen, Q.R.; Briggs, J.; Cassavaugh, J.; Srinivasan, S.; Lizardo, M.M.; Mendoza, A.; Xia, A.Y.; Avadhani, N.; et al. Dysregulation of ezrin phosphorylation prevents metastasis and alters cellular metabolism in osteosarcoma. Cancer Res. 2012, 72, 1001–1012. [Google Scholar] [CrossRef] [Green Version]
  196. Sarrio, D.; Rodriguez-Pinilla, S.M.; Dotor, A.; Calero, F.; Hardisson, D.; Palacios, J. Abnormal ezrin localization is associated with clinicopathological features in invasive breast carcinomas. Breast Cancer Res. Treat. 2006, 98, 71–79. [Google Scholar] [CrossRef]
  197. Barik, G.K.; Sahay, O.; Paul, D.; Santra, M.K. Ezrin gone rogue in cancer progression and metastasis: An enticing therapeutic target. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188753. [Google Scholar] [CrossRef]
  198. Ma, L.; Jiang, T. Clinical implications of Ezrin and CD44 coexpression in breast cancer. Oncol. Rep. 2013, 30, 1899–1905. [Google Scholar] [CrossRef] [Green Version]
  199. Penchev, V.R.; Chang, Y.T.; Begum, A.; Ewachiw, T.; Gocke, C.; Li, J.; McMillan, R.H.; Wang, Q.; Anders, R.; Marchionni, L.; et al. Ezrin Promotes Stem Cell Properties in Pancreatic Ductal Adenocarcinoma. Mol. Cancer Res. 2019, 17, 929–936. [Google Scholar] [CrossRef]
  200. Hoskin, V.; Szeto, A.; Ghaffari, A.; Greer, P.A.; Cote, G.P.; Elliott, B.E. Ezrin regulates focal adhesion and invadopodia dynamics by altering calpain activity to promote breast cancer cell invasion. Mol. Biol. Cell 2015, 26, 3464–3479. [Google Scholar] [CrossRef]
  201. McBeath, R.; Pirone, D.M.; Nelson, C.M.; Bhadriraju, K.; Chen, C.S. Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev. Cell 2004, 6, 483–495. [Google Scholar] [CrossRef] [Green Version]
  202. Sun, X.; Li, K.; Hase, M.; Zha, R.; Feng, Y.; Li, B.Y.; Yokota, H. Suppression of breast cancer-associated bone loss with osteoblast proteomes via Hsp90ab1/moesin-mediated inhibition of TGFbeta/FN1/CD44 signaling. Theranostics 2022, 12, 929–943. [Google Scholar] [CrossRef]
  203. Zhu, X.; Morales, F.C.; Agarwal, N.K.; Dogruluk, T.; Gagea, M.; Georgescu, M.M. Moesin is a glioma progression marker that induces proliferation and Wnt/beta-catenin pathway activation via interaction with CD44. Cancer Res. 2013, 73, 1142–1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Hong, A.W.; Meng, Z.; Plouffe, S.W.; Lin, Z.; Zhang, M.; Guan, K.L. Critical roles of phosphoinositides and NF2 in Hippo pathway regulation. Genes Dev. 2020, 34, 511–525. [Google Scholar] [CrossRef] [PubMed]
  205. Tian, Q.; Gao, H.; Zhou, Y.; Zhu, L.; Yang, J.; Wang, B.; Liu, P.; Yang, J. RICH1 inhibits breast cancer stem cell traits through activating kinases cascade of Hippo signaling by competing with Merlin for binding to Amot-p80. Cell Death Dis. 2022, 13, 71. [Google Scholar] [CrossRef] [PubMed]
  206. Cordenonsi, M.; Zanconato, F.; Azzolin, L.; Forcato, M.; Rosato, A.; Frasson, C.; Inui, M.; Montagner, M.; Parenti, A.R.; Poletti, A.; et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell 2011, 147, 759–772. [Google Scholar] [CrossRef]
  207. Marino, N.; Marshall, J.C.; Collins, J.W.; Zhou, M.; Qian, Y.; Veenstra, T.; Steeg, P.S. Nm23-h1 binds to gelsolin and inactivates its actin-severing capacity to promote tumor cell motility and metastasis. Cancer Res. 2013, 73, 5949–5962. [Google Scholar] [CrossRef] [Green Version]
  208. Chen, Z.Y.; Wang, P.W.; Shieh, D.B.; Chiu, K.Y.; Liou, Y.M. Involvement of gelsolin in TGF-beta 1 induced epithelial to mesenchymal transition in breast cancer cells. J. Biomed. Sci. 2015, 22, 90. [Google Scholar] [CrossRef] [Green Version]
  209. Liao, P.H.; Hsu, H.H.; Chen, T.S.; Chen, M.C.; Day, C.H.; Tu, C.C.; Lin, Y.M.; Tsai, F.J.; Kuo, W.W.; Huang, C.Y. Phosphorylation of cofilin-1 by ERK confers HDAC inhibitor resistance in hepatocellular carcinoma cells via decreased ROS-mediated mitochondria injury. Oncogene 2017, 36, 1978–1990. [Google Scholar] [CrossRef]
  210. Lu, L.I.; Fu, N.I.; Luo, X.U.; Li, X.Y.; Li, X.P. Overexpression of cofilin 1 in prostate cancer and the corresponding clinical implications. Oncol. Lett. 2015, 9, 2757–2761. [Google Scholar] [CrossRef]
  211. Ozeki, M.; Aini, W.; Miyagawa-Hayashino, A.; Tamaki, K. Prevention of Cell Growth by Suppression of Villin Expression in Lithocholic Acid-Stimulated HepG2 Cells. J. Histochem. Cytochem. 2019, 67, 129–141. [Google Scholar] [CrossRef] [PubMed]
  212. Qiao, X.T.; Gumucio, D.L. Current molecular markers for gastric progenitor cells and gastric cancer stem cells. J. Gastroenterol. 2011, 46, 855–865. [Google Scholar] [CrossRef] [PubMed]
  213. Wang, C.X.; Liu, B.; Wang, Y.F.; Zhang, R.S.; Yu, B.; Lu, Z.F.; Shi, Q.L.; Zhou, X.J. Pulmonary enteric adenocarcinoma: A study of the clinicopathologic and molecular status of nine cases. Int. J. Clin. Exp. Pathol. 2014, 7, 1266–1274. [Google Scholar] [PubMed]
  214. Zheng, H.; Yang, Y.; Hong, Y.G.; Wang, M.C.; Yuan, S.X.; Wang, Z.G.; Bi, F.R.; Hao, L.Q.; Yan, H.L.; Zhou, W.P. Tropomodulin 3 modulates EGFR-PI3K-AKT signaling to drive hepatocellular carcinoma metastasis. Mol. Carcinog. 2019, 58, 1897–1907. [Google Scholar] [CrossRef] [PubMed]
  215. Ito-Kureha, T.; Koshikawa, N.; Yamamoto, M.; Semba, K.; Yamaguchi, N.; Yamamoto, T.; Seiki, M.; Inoue, J. Tropomodulin 1 expression driven by NF-kappaB enhances breast cancer growth. Cancer Res. 2015, 75, 62–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Xiang, X.; Wang, Y.; Zhang, H.; Piao, J.; Muthusamy, S.; Wang, L.; Deng, Y.; Zhang, W.; Kuang, R.; Billadeau, D.D.; et al. Vasodilator-stimulated phosphoprotein promotes liver metastasis of gastrointestinal cancer by activating a beta1-integrin-FAK-YAP1/TAZ signaling pathway. NPJ Precis. Oncol. 2018, 2, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Chen, H.; Dai, G.; Cai, Y.; Gong, Q.; Wu, W.; Gao, M.; Fei, Z. Vasodilator-stimulated phosphoprotein (VASP), a novel target of miR-4455, promotes gastric cancer cell proliferation, migration, and invasion, through activating the PI3K/AKT signaling pathway. Cancer Cell. Int. 2018, 18, 97. [Google Scholar] [CrossRef]
  218. Dybdal-Hargreaves, N.F.; Risinger, A.L.; Mooberry, S.L. Regulation of E-cadherin localization by microtubule targeting agents: Rapid promotion of cortical E-cadherin through p130Cas/Src inhibition by eribulin. Oncotarget 2018, 9, 5545–5561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Whipple, R.A.; Matrone, M.A.; Cho, E.H.; Balzer, E.M.; Vitolo, M.I.; Yoon, J.R.; Ioffe, O.B.; Tuttle, K.C.; Yang, J.; Martin, S.S. Epithelial-to-mesenchymal transition promotes tubulin detyrosination and microtentacles that enhance endothelial engagement. Cancer Res. 2010, 70, 8127–8137. [Google Scholar] [CrossRef] [Green Version]
  220. Ceccarelli, M.; Barthel, F.P.; Malta, T.M.; Sabedot, T.S.; Salama, S.R.; Murray, B.A.; Morozova, O.; Newton, Y.; Radenbaugh, A.; Pagnotta, S.M.; et al. Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma. Cell 2016, 164, 550–563. [Google Scholar] [CrossRef]
  221. Bonneau, C.; Gurard-Levin, Z.A.; Andre, F.; Pusztai, L.; Rouzier, R. Predictive and Prognostic Value of the TauProtein in Breast Cancer. Anticancer Res. 2015, 35, 5179–5184. [Google Scholar] [PubMed]
  222. Correa-Saez, A.; Jimenez-Izquierdo, R.; Garrido-Rodriguez, M.; Morrugares, R.; Munoz, E.; Calzado, M.A. Updating dual-specificity tyrosine-phosphorylation-regulated kinase 2 (DYRK2): Molecular basis, functions and role in diseases. Cell Mol. Life Sci. 2020, 77, 4747–4763. [Google Scholar] [CrossRef] [PubMed]
  223. Kuo, T.C.; Li, L.W.; Pan, S.H.; Fang, J.M.; Liu, J.H.; Cheng, T.J.; Wang, C.J.; Hung, P.F.; Chen, H.Y.; Hong, T.M.; et al. Purine-Type Compounds Induce Microtubule Fragmentation and Lung Cancer Cell Death through Interaction with Katanin. J. Med. Chem. 2016, 59, 8521–8534. [Google Scholar] [CrossRef]
  224. Wang, L.; Tantai, J.; Zhu, X. Katanin P60: A potential biomarker for lymph node metastasis and prognosis for non-small cell lung cancer. World J. Surg. Oncol. 2020, 18, 157. [Google Scholar] [CrossRef]
  225. Zhao, T.; He, Q.; Liu, Z.; Ding, X.; Zhou, X.; Wang, A. Angiotensin II type 2 receptor-interacting protein 3a suppresses proliferation, migration and invasion in tongue squamous cell carcinoma via the extracellular signal-regulated kinase-Snai2 pathway. Oncol. Lett. 2016, 11, 340–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Liu, B.; Zhang, G.; Cui, S.; Du, G. Upregulation of KIF11 in TP53 Mutant Glioma Promotes Tumor Stemness and Drug Resistance. Cell Mol. Neurobiol. 2022, 42, 1477–1485. [Google Scholar] [CrossRef] [PubMed]
  227. Li, N.; Jiang, P.; Du, W.; Wu, Z.; Li, C.; Qiao, M.; Yang, X.; Wu, M. Siva1 suppresses epithelial-mesenchymal transition and metastasis of tumor cells by inhibiting stathmin and stabilizing microtubules. Proc. Natl. Acad. Sci. USA 2011, 108, 12851–12856. [Google Scholar] [CrossRef] [Green Version]
  228. Christofori, G. New signals from the invasive front. Nature 2006, 441, 444–450. [Google Scholar] [CrossRef]
  229. Pongrakhananon, V.; Wattanathamsan, O.; Takeichi, M.; Chetprayoon, P.; Chanvorachote, P. Loss of CAMSAP3 promotes EMT via the modification of microtubule-Akt machinery. J. Cell Sci. 2018, 131, jcs216168. [Google Scholar] [CrossRef] [Green Version]
  230. Harada, T.; Swift, J.; Irianto, J.; Shin, J.W.; Spinler, K.R.; Athirasala, A.; Diegmiller, R.; Dingal, P.C.; Ivanovska, I.L.; Discher, D.E. Nuclear lamin stiffness is a barrier to 3D migration, but softness can limit survival. J. Cell Biol. 2014, 204, 669–682. [Google Scholar] [CrossRef]
  231. Thomas, D.; Thiagarajan, P.S.; Rai, V.; Reizes, O.; Lathia, J.; Egelhoff, T. Increased cancer stem cell invasion is mediated by myosin IIB and nuclear translocation. Oncotarget 2016, 7, 47586–47592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Thomas, D.G.; Yenepalli, A.; Denais, C.M.; Rape, A.; Beach, J.R.; Wang, Y.L.; Schiemann, W.P.; Baskaran, H.; Lammerding, J.; Egelhoff, T.T. Non-muscle myosin IIB is critical for nuclear translocation during 3D invasion. J. Cell Biol. 2015, 210, 583–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Derycke, L.; Stove, C.; Vercoutter-Edouart, A.S.; De Wever, O.; Dolle, L.; Colpaert, N.; Depypere, H.; Michalski, J.C.; Bracke, M. The role of non-muscle myosin IIA in aggregation and invasion of human MCF-7 breast cancer cells. Int. J. Dev. Biol. 2011, 55, 835–840. [Google Scholar] [CrossRef]
  234. Beach, J.R.; Hussey, G.S.; Miller, T.E.; Chaudhury, A.; Patel, P.; Monslow, J.; Zheng, Q.; Keri, R.A.; Reizes, O.; Bresnick, A.R.; et al. Myosin II isoform switching mediates invasiveness after TGF-beta-induced epithelial-mesenchymal transition. Proc. Natl. Acad. Sci. USA 2011, 108, 17991–17996. [Google Scholar] [CrossRef] [Green Version]
  235. Wu, S.; Du, Y.; Beckford, J.; Alachkar, H. Upregulation of the EMT marker vimentin is associated with poor clinical outcome in acute myeloid leukemia. J. Transl. Med. 2018, 16, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Vuoriluoto, K.; Haugen, H.; Kiviluoto, S.; Mpindi, J.P.; Nevo, J.; Gjerdrum, C.; Tiron, C.; Lorens, J.B.; Ivaska, J. Vimentin regulates EMT induction by Slug and oncogenic H-Ras and migration by governing Axl expression in breast cancer. Oncogene 2011, 30, 1436–1448. [Google Scholar] [CrossRef] [Green Version]
  237. Usman, S.; Waseem, N.H.; Nguyen, T.K.N.; Mohsin, S.; Jamal, A.; Teh, M.T.; Waseem, A. Vimentin Is at the Heart of Epithelial Mesenchymal Transition (EMT) Mediated Metastasis. Cancers 2021, 13, 4985. [Google Scholar] [CrossRef]
  238. Neradil, J.; Veselska, R. Nestin as a marker of cancer stem cells. Cancer Sci. 2015, 106, 803–811. [Google Scholar] [CrossRef] [Green Version]
  239. Matsuda, Y.; Hagio, M.; Ishiwata, T. Nestin: A novel angiogenesis marker and possible target for tumor angiogenesis. World J. Gastroenterol. 2013, 19, 42–48. [Google Scholar] [CrossRef]
  240. Zhao, Z.; Lu, P.; Zhang, H.; Xu, H.; Gao, N.; Li, M.; Liu, C. Nestin positively regulates the Wnt/beta-catenin pathway and the proliferation, survival and invasiveness of breast cancer stem cells. Breast Cancer Res. 2014, 16, 408. [Google Scholar] [CrossRef]
  241. Tsai, F.J.; Lai, M.T.; Cheng, J.; Chao, S.C.; Korla, P.K.; Chen, H.J.; Lin, C.M.; Tsai, M.H.; Hua, C.H.; Jan, C.I.; et al. Novel K6-K14 keratin fusion enhances cancer stemness and aggressiveness in oral squamous cell carcinoma. Oncogene 2019, 38, 5113–5126. [Google Scholar] [CrossRef] [PubMed]
  242. Huang, W.C.; Jang, T.H.; Tung, S.L.; Yen, T.C.; Chan, S.H.; Wang, L.H. A novel miR-365-3p/EHF/keratin 16 axis promotes oral squamous cell carcinoma metastasis, cancer stemness and drug resistance via enhancing beta5-integrin/c-met signaling pathway. J. Exp. Clin. Cancer Res. 2019, 38, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Jang, T.H.; Huang, W.C.; Tung, S.L.; Lin, S.C.; Chen, P.M.; Cho, C.Y.; Yang, Y.Y.; Yen, T.C.; Lo, G.H.; Chuang, S.E.; et al. MicroRNA-485-5p targets keratin 17 to regulate oral cancer stemness and chemoresistance via the integrin/FAK/Src/ERK/beta-catenin pathway. J. Biomed. Sci. 2022, 29, 42. [Google Scholar] [CrossRef]
  244. Kim, S.Y. Cancer Energy Metabolism: Shutting Power off Cancer Factory. Biomol. Ther. 2018, 26, 39–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Hu, H.; Juvekar, A.; Lyssiotis, C.A.; Lien, E.C.; Albeck, J.G.; Oh, D.; Varma, G.; Hung, Y.P.; Ullas, S.; Lauring, J.; et al. Phosphoinositide 3-Kinase Regulates Glycolysis through Mobilization of Aldolase from the Actin Cytoskeleton. Cell 2016, 164, 433–446. [Google Scholar] [CrossRef] [Green Version]
  246. Shankar, J.; Nabi, I.R. Actin cytoskeleton regulation of epithelial mesenchymal transition in metastatic cancer cells. PLoS ONE 2015, 10, e0119954. [Google Scholar] [CrossRef] [Green Version]
  247. Zhao, J.; Zhang, J.; Yu, M.; Xie, Y.; Huang, Y.; Wolff, D.W.; Abel, P.W.; Tu, Y. Mitochondrial dynamics regulates migration and invasion of breast cancer cells. Oncogene 2013, 32, 4814–4824. [Google Scholar] [CrossRef]
  248. Taulet, N.; Delorme-Walker, V.D.; DerMardirossian, C. Reactive oxygen species regulate protrusion efficiency by controlling actin dynamics. PLoS ONE 2012, 7, e41342. [Google Scholar] [CrossRef]
  249. Bonello, T.T.; Stehn, J.R.; Gunning, P.W. New approaches to targeting the actin cytoskeleton for chemotherapy. Future Med. Chem. 2009, 1, 1311–1331. [Google Scholar] [CrossRef]
  250. Guerriero, C.J.; Weisz, O.A. N-WASP inhibitor wiskostatin nonselectively perturbs membrane transport by decreasing cellular ATP levels. Am. J. Physiol. Cell Physiol. 2007, 292, C1562–C1566. [Google Scholar] [CrossRef]
  251. Hetrick, B.; Han, M.S.; Helgeson, L.A.; Nolen, B.J. Small molecules CK-666 and CK-869 inhibit actin-related protein 2/3 complex by blocking an activating conformational change. Chem. Biol. 2013, 20, 701–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Peterson, J.R.; Mitchison, T.J. Small molecules, big impact: A history of chemical inhibitors and the cytoskeleton. Chem. Biol. 2002, 9, 1275–1285. [Google Scholar] [CrossRef] [Green Version]
  253. Kim, H.C.; Jo, Y.J.; Kim, N.H.; Namgoong, S. Small molecule inhibitor of formin homology 2 domains (SMIFH2) reveals the roles of the formin family of proteins in spindle assembly and asymmetric division in mouse oocytes. PLoS ONE 2015, 10, e0123438. [Google Scholar] [CrossRef] [Green Version]
  254. Ziske, M.A.; Pettee, K.M.; Khaing, M.; Rubinic, K.; Eisenmann, K.M. SMIFH2-mediated mDia formin functional inhibition potentiates chemotherapeutic targeting of human ovarian cancer spheroids. Biochem. Biophys. Res. Commun. 2016, 472, 33–39. [Google Scholar] [CrossRef] [PubMed]
  255. Orgaz, J.L.; Herraiz, C.; Sanz-Moreno, V. Rho GTPases modulate malignant transformation of tumor cells. Small GTPases 2014, 5, e29019. [Google Scholar] [CrossRef]
  256. Biber, G.; Ben-Shmuel, A.; Sabag, B.; Barda-Saad, M. Actin regulators in cancer progression and metastases: From structure and function to cytoskeletal dynamics. Int. Rev. Cell Mol. Biol. 2020, 356, 131–196. [Google Scholar] [CrossRef]
  257. Arias-Romero, L.E.; Chernoff, J. Targeting Cdc42 in cancer. Expert Opin. Ther. Targets 2013, 17, 1263–1273. [Google Scholar] [CrossRef] [Green Version]
  258. Guo, Y.; Kenney, S.R.; Muller, C.Y.; Adams, S.; Rutledge, T.; Romero, E.; Murray-Krezan, C.; Prekeris, R.; Sklar, L.A.; Hudson, L.G.; et al. R-Ketorolac Targets Cdc42 and Rac1 and Alters Ovarian Cancer Cell Behaviors Critical for Invasion and Metastasis. Mol. Cancer Ther. 2015, 14, 2215–2227. [Google Scholar] [CrossRef] [Green Version]
  259. Humphries-Bickley, T.; Castillo-Pichardo, L.; Hernandez-O’Farrill, E.; Borrero-Garcia, L.D.; Forestier-Roman, I.; Gerena, Y.; Blanco, M.; Rivera-Robles, M.J.; Rodriguez-Medina, J.R.; Cubano, L.A.; et al. Characterization of a Dual Rac/Cdc42 Inhibitor MBQ-167 in Metastatic Cancer. Mol. Cancer Ther. 2017, 16, 805–818. [Google Scholar] [CrossRef] [Green Version]
  260. Maldonado, M.D.M.; Dharmawardhane, S. Targeting Rac and Cdc42 GTPases in Cancer. Cancer Res. 2018, 78, 3101–3111. [Google Scholar] [CrossRef]
  261. Schofield, A.V.; Bernard, O. Rho-associated coiled-coil kinase (ROCK) signaling and disease. Crit. Rev. Biochem. Mol. Biol. 2013, 48, 301–316. [Google Scholar] [CrossRef] [PubMed]
  262. Shahbazi, R.; Baradaran, B.; Khordadmehr, M.; Safaei, S.; Baghbanzadeh, A.; Jigari, F.; Ezzati, H. Targeting ROCK signaling in health, malignant and non-malignant diseases. Immunol. Lett. 2020, 219, 15–26. [Google Scholar] [CrossRef] [PubMed]
  263. Yang, S.; Kim, H.M. ROCK inhibition activates MCF-7 cells. PLoS ONE 2014, 9, e88489. [Google Scholar] [CrossRef] [PubMed]
  264. Lock, F.E.; Ryan, K.R.; Poulter, N.S.; Parsons, M.; Hotchin, N.A. Differential regulation of adhesion complex turnover by ROCK1 and ROCK2. PLoS ONE 2012, 7, e31423. [Google Scholar] [CrossRef] [Green Version]
  265. Wei, L.; Surma, M.; Shi, S.; Lambert-Cheatham, N.; Shi, J. Novel Insights into the Roles of Rho Kinase in Cancer. Arch. Immunol. Ther. Exp. 2016, 64, 259–278. [Google Scholar] [CrossRef] [Green Version]
  266. Pimm, M.L.; Hotaling, J.; Henty-Ridilla, J.L. Profilin choreographs actin and microtubules in cells and cancer. Int. Rev. Cell Mol. Biol. 2020, 355, 155–204. [Google Scholar] [CrossRef]
  267. Gau, D.; Lewis, T.; McDermott, L.; Wipf, P.; Koes, D.; Roy, P. Structure-based virtual screening identifies a small-molecule inhibitor of the profilin 1-actin interaction. J. Biol. Chem. 2018, 293, 2606–2616. [Google Scholar] [CrossRef] [Green Version]
  268. Huang, D.; Wang, S.; Wang, A.; Chen, X.; Zhang, H. Thymosin beta 4 silencing suppresses proliferation and invasion of non-small cell lung cancer cells by repressing Notch1 activation. Acta Biochim. Biophys. Sin. 2016, 48, 788–794. [Google Scholar] [CrossRef] [Green Version]
  269. Ristic, B.; Kopel, J.; Sherazi, S.A.A.; Gupta, S.; Sachdeva, S.; Bansal, P.; Ali, A.; Perisetti, A.; Goyal, H. Emerging Role of Fascin-1 in the Pathogenesis, Diagnosis, and Treatment of the Gastrointestinal Cancers. Cancers 2021, 13, 2536. [Google Scholar] [CrossRef]
  270. Montoro-Garcia, S.; Alburquerque-Gonzalez, B.; Bernabe-Garcia, A.; Bernabe-Garcia, M.; Rodrigues, P.C.; den-Haan, H.; Luque, I.; Nicolas, F.J.; Perez-Sanchez, H.; Cayuela, M.L.; et al. Novel anti-invasive properties of a Fascin1 inhibitor on colorectal cancer cells. J. Mol. Med. 2020, 98, 383–394. [Google Scholar] [CrossRef]
  271. Mahmoud, A.; Elkhalifa, D.; Alali, F.; Al Moustafa, A.E.; Khalil, A. Novel Polymethoxylated Chalcones as Potential Compounds Against KRAS-Mutant Colorectal Cancers. Curr. Pharm. Des. 2020, 26, 1622–1633. [Google Scholar] [CrossRef] [PubMed]
  272. Huang, J.; Dey, R.; Wang, Y.; Jakoncic, J.; Kurinov, I.; Huang, X.Y. Structural Insights into the Induced-fit Inhibition of Fascin by a Small-Molecule Inhibitor. J. Mol. Biol. 2018, 430, 1324–1335. [Google Scholar] [CrossRef] [PubMed]
  273. Chung, V.; Jhaveri, K.L.; Hoff, D.D.V.; Huang, X.-Y.; Garmey, E.G.; Zhang, J.; Tsai, F.Y.-C. Phase 1A clinical trial of the first-in-class fascin inhibitor NP-G2-044 evaluating safety and anti-tumor activity in patients with advanced and metastatic solid tumors. J. Clin. Oncol. 2021, 39, 2548. [Google Scholar] [CrossRef]
  274. Alburquerque-Gonzalez, B.; Bernabe-Garcia, A.; Bernabe-Garcia, M.; Ruiz-Sanz, J.; Lopez-Calderon, F.F.; Gonnelli, L.; Banci, L.; Pena-Garcia, J.; Luque, I.; Nicolas, F.J.; et al. The FDA-Approved Antiviral Raltegravir Inhibits Fascin1-Dependent Invasion of Colorectal Tumor Cells In Vitro and In Vivo. Cancers 2021, 13, 861. [Google Scholar] [CrossRef]
  275. Schenk, M.; Aykut, B.; Teske, C.; Giese, N.A.; Weitz, J.; Welsch, T. Salinomycin inhibits growth of pancreatic cancer and cancer cell migration by disruption of actin stress fiber integrity. Cancer Lett. 2015, 358, 161–169. [Google Scholar] [CrossRef]
  276. Alburquerque-Gonzalez, B.; Bernabe-Garcia, M.; Montoro-Garcia, S.; Bernabe-Garcia, A.; Rodrigues, P.C.; Ruiz Sanz, J.; Lopez-Calderon, F.F.; Luque, I.; Nicolas, F.J.; Cayuela, M.L.; et al. New role of the antidepressant imipramine as a Fascin1 inhibitor in colorectal cancer cells. Exp. Mol. Med. 2020, 52, 281–292. [Google Scholar] [CrossRef] [Green Version]
  277. Bulut, G.; Hong, S.H.; Chen, K.; Beauchamp, E.M.; Rahim, S.; Kosturko, G.W.; Glasgow, E.; Dakshanamurthy, S.; Lee, H.S.; Daar, I.; et al. Small molecule inhibitors of ezrin inhibit the invasive phenotype of osteosarcoma cells. Oncogene 2012, 31, 269–281. [Google Scholar] [CrossRef] [Green Version]
  278. Celik, H.; Bulut, G.; Han, J.; Graham, G.T.; Minas, T.Z.; Conn, E.J.; Hong, S.H.; Pauly, G.T.; Hayran, M.; Li, X.; et al. Ezrin Inhibition Up-regulates Stress Response Gene Expression. J. Biol. Chem. 2016, 291, 13257–13270. [Google Scholar] [CrossRef] [Green Version]
  279. Jeong, J.; Choi, J.; Kim, W.; Dann, P.; Takyar, F.; Gefter, J.V.; Friedman, P.A.; Wysolmerski, J.J. Inhibition of ezrin causes PKCalpha-mediated internalization of erbb2/HER2 tyrosine kinase in breast cancer cells. J. Biol. Chem. 2019, 294, 887–901. [Google Scholar] [CrossRef] [Green Version]
  280. Gavert, N.; Ben-Shmuel, A.; Lemmon, V.; Brabletz, T.; Ben-Ze’ev, A. Nuclear factor-kappaB signaling and ezrin are essential for L1-mediated metastasis of colon cancer cells. J. Cell Sci. 2010, 123, 2135–2143. [Google Scholar] [CrossRef]
  281. Lugini, L.; Matarrese, P.; Tinari, A.; Lozupone, F.; Federici, C.; Iessi, E.; Gentile, M.; Luciani, F.; Parmiani, G.; Rivoltini, L.; et al. Cannibalism of live lymphocytes by human metastatic but not primary melanoma cells. Cancer Res. 2006, 66, 3629–3638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Shang, X.; Wang, Y.; Zhao, Q.; Wu, K.; Li, X.; Ji, X.; He, R.; Zhang, W. siRNAs target sites selection of ezrin and the influence of RNA interference on ezrin expression and biological characters of osteosarcoma cells. Mol. Cell Biochem. 2012, 364, 363–371. [Google Scholar] [CrossRef] [PubMed]
  283. Pignochino, Y.; Grignani, G.; Cavalloni, G.; Motta, M.; Tapparo, M.; Bruno, S.; Bottos, A.; Gammaitoni, L.; Migliardi, G.; Camussi, G.; et al. Sorafenib blocks tumour growth, angiogenesis and metastatic potential in preclinical models of osteosarcoma through a mechanism potentially involving the inhibition of ERK1/2, MCL-1 and ezrin pathways. Mol. Cancer 2009, 8, 118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Chen, Z.; Hou, R.; Gao, S.; Song, D.; Feng, Y. Baicalein Inhibits Proliferation Activity of Human Colorectal Cancer Cells HCT116 Through Downregulation of Ezrin. Cell Physiol. Biochem. 2018, 49, 2035–2046. [Google Scholar] [CrossRef]
  285. Zhang, J.; Yang, W.; Zhou, Y.B.; Xiang, Y.X.; Wang, L.S.; Hu, W.K.; Wang, W.J. Baicalein inhibits osteosarcoma cell proliferation and invasion through the miR183/Ezrin pathway. Mol. Med. Rep. 2018, 18, 1104–1112. [Google Scholar] [CrossRef] [Green Version]
  286. Mukhtar, E.; Adhami, V.M.; Mukhtar, H. Targeting microtubules by natural agents for cancer therapy. Mol. Cancer Ther. 2014, 13, 275–284. [Google Scholar] [CrossRef] [Green Version]
  287. Weaver, B.A. How Taxol/paclitaxel kills cancer cells. Mol. Biol. Cell 2014, 25, 2677–2681. [Google Scholar] [CrossRef]
  288. Abu Samaan, T.M.; Samec, M.; Liskova, A.; Kubatka, P.; Busselberg, D. Paclitaxel’s Mechanistic and Clinical Effects on Breast Cancer. Biomolecules 2019, 9, 789. [Google Scholar] [CrossRef] [Green Version]
  289. Sharma, P.; Alsharif, S.; Fallatah, A.; Chung, B.M. Intermediate Filaments as Effectors of Cancer Development and Metastasis: A Focus on Keratins, Vimentin, and Nestin. Cells 2019, 8, 497. [Google Scholar] [CrossRef] [Green Version]
  290. Lee, I.C.; Choi, B.Y. Withaferin-A--A Natural Anticancer Agent with Pleitropic Mechanisms of Action. Int. J. Mol. Sci. 2016, 17, 290. [Google Scholar] [CrossRef]
  291. Nagalingam, A.; Kuppusamy, P.; Singh, S.V.; Sharma, D.; Saxena, N.K. Mechanistic elucidation of the antitumor properties of withaferin a in breast cancer. Cancer Res. 2014, 74, 2617–2629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Bhat, K.M.; Setaluri, V. Microtubule-associated proteins as targets in cancer chemotherapy. Clin. Cancer Res. 2007, 13, 2849–2854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Smoter, M.; Bodnar, L.; Duchnowska, R.; Stec, R.; Grala, B.; Szczylik, C. The role of Tau protein in resistance to paclitaxel. Cancer Chemother. Pharmacol. 2011, 68, 553–557. [Google Scholar] [CrossRef] [Green Version]
  294. Kavallaris, M. Microtubules and resistance to tubulin-binding agents. Nat. Rev. Cancer 2010, 10, 194–204. [Google Scholar] [CrossRef]
  295. Sakowicz, R.; Finer, J.T.; Beraud, C.; Crompton, A.; Lewis, E.; Fritsch, A.; Lee, Y.; Mak, J.; Moody, R.; Turincio, R.; et al. Antitumor activity of a kinesin inhibitor. Cancer Res. 2004, 64, 3276–3280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Gomez, H.L.; Philco, M.; Pimentel, P.; Kiyan, M.; Monsalvo, M.L.; Conlan, M.G.; Saikali, K.G.; Chen, M.M.; Seroogy, J.J.; Wolff, A.A.; et al. Phase I dose-escalation and pharmacokinetic study of ispinesib, a kinesin spindle protein inhibitor, administered on days 1 and 15 of a 28-day schedule in patients with no prior treatment for advanced breast cancer. Anticancer Drugs 2012, 23, 335–341. [Google Scholar] [CrossRef]
  297. Holen, K.D.; Belani, C.P.; Wilding, G.; Ramalingam, S.; Volkman, J.L.; Ramanathan, R.K.; Vasist, L.S.; Bowen, C.J.; Hodge, J.P.; Dar, M.M.; et al. A first in human study of SB-743921, a kinesin spindle protein inhibitor, to determine pharmacokinetics, biologic effects and establish a recommended phase II dose. Cancer Chemother. Pharmacol. 2011, 67, 447–454. [Google Scholar] [CrossRef] [Green Version]
  298. Lucanus, A.J.; Yip, G.W. Kinesin superfamily: Roles in breast cancer, patient prognosis and therapeutics. Oncogene 2018, 37, 833–838. [Google Scholar] [CrossRef]
  299. Belletti, B.; Baldassarre, G. Stathmin: A protein with many tasks. New biomarker and potential target in cancer. Expert Opin. Ther. Targets 2011, 15, 1249–1266. [Google Scholar] [CrossRef]
  300. Miceli, C.; Tejada, A.; Castaneda, A.; Mistry, S.J. Cell cycle inhibition therapy that targets stathmin in in vitro and in vivo models of breast cancer. Cancer Gene Ther. 2013, 20, 298–307. [Google Scholar] [CrossRef]
  301. Long, M.; Yin, G.; Liu, L.; Lin, F.; Wang, X.; Ren, J.; Wei, J.; Dong, K.; Zhang, H. Adenovirus-mediated Aurora A shRNA driven by stathmin promoter suppressed tumor growth and enhanced paclitaxel chemotherapy sensitivity in human breast carcinoma cells. Cancer Gene Ther. 2012, 19, 271–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  302. Chen, H.; Chomyn, A.; Chan, D.C. Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J. Biol. Chem. 2005, 280, 26185–26192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Ferreira, J.C.B.; Campos, J.C.; Qvit, N.; Qi, X.; Bozi, L.H.M.; Bechara, L.R.G.; Lima, V.M.; Queliconi, B.B.; Disatnik, M.H.; Dourado, P.M.M.; et al. A selective inhibitor of mitofusin 1-betaIIPKC association improves heart failure outcome in rats. Nat. Commun. 2019, 10, 329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Lonardo, E.; Cioffi, M.; Sancho, P.; Sanchez-Ripoll, Y.; Trabulo, S.M.; Dorado, J.; Balic, A.; Hidalgo, M.; Heeschen, C. Metformin targets the metabolic achilles heel of human pancreatic cancer stem cells. PLoS ONE 2013, 8, e76518. [Google Scholar] [CrossRef] [Green Version]
  305. Wheaton, W.W.; Weinberg, S.E.; Hamanaka, R.B.; Soberanes, S.; Sullivan, L.B.; Anso, E.; Glasauer, A.; Dufour, E.; Mutlu, G.M.; Budigner, G.S.; et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. Elife 2014, 3, e02242. [Google Scholar] [CrossRef]
  306. Choi, J.; Lee, J.H.; Koh, I.; Shim, J.K.; Park, J.; Jeon, J.Y.; Yun, M.; Kim, S.H.; Yook, J.I.; Kim, E.H.; et al. Inhibiting stemness and invasive properties of glioblastoma tumorsphere by combined treatment with temozolomide and a newly designed biguanide (HL156A). Oncotarget 2016, 7, 65643–65659. [Google Scholar] [CrossRef]
Figure 1. Actin-binding proteins. This figure shows the proteins involved in actin binding, capping, cross-linking, bundling, severing and anchoring.
Figure 1. Actin-binding proteins. This figure shows the proteins involved in actin binding, capping, cross-linking, bundling, severing and anchoring.
Pharmaceuticals 15 01369 g001
Figure 2. Schematic representation of microtubule−associated protein (MAP) binding to microtubules at different sites. The main proteins include microtubule crystal−binding proteins (MAP tau, MAP2, ATIP3, and katanin), microtubule movement proteins in which kinesins carry organelles or molecules to the plus−end of microtubules (anterograde transport); dyneins that transport cellular molecules to the minus−end of microtubules (retrograde transport); multisite microtubule−binding protein stathmin (STMN1); microtubule plus−end−binding proteins (EB and CLASPs); and microtubule minus−end-binding proteins (CAMSAP1, CAMSAP2 and CAMSAP3).
Figure 2. Schematic representation of microtubule−associated protein (MAP) binding to microtubules at different sites. The main proteins include microtubule crystal−binding proteins (MAP tau, MAP2, ATIP3, and katanin), microtubule movement proteins in which kinesins carry organelles or molecules to the plus−end of microtubules (anterograde transport); dyneins that transport cellular molecules to the minus−end of microtubules (retrograde transport); multisite microtubule−binding protein stathmin (STMN1); microtubule plus−end−binding proteins (EB and CLASPs); and microtubule minus−end-binding proteins (CAMSAP1, CAMSAP2 and CAMSAP3).
Pharmaceuticals 15 01369 g002
Table 1. Mechanism of cytoskeleton and cytoskeleton-related proteins on CSCs.
Table 1. Mechanism of cytoskeleton and cytoskeleton-related proteins on CSCs.
ClassProteinsMechanismEffect
ActinActivation of the downstream Rho/ROCK pathway via YAP/TAZFacilitating the survival of CSCs
Monomer-binding proteinsProfilinDirect regulation of stem cell-associated transcription factorsMaintaining the stemness of CSCs
Thymosin β4Activation of the BMP pathway, followed by JNK activation via the TAB1 and TAK1 complexesMaintaining the stemness of CSCs
TwinfilinEnhancing the activity of the MKL1 and actin cytoskeleton dynamicsFacilitating the survival of CSCs
Cross-linking and bundling proteinsFascinActivation of β-catenin protein signaling via FAKPromoting CSC function
FilamininInteracts with Rho GTPases that activate cell migration and Ras GTPases that inhibit cell migrationPromoting CSC function
SpectrinInhibition of CSCs by β-catenin-induced differentiationInhibiting the properties of CSCs
α actininActs through the Akt/GSK-3β/β-catenin axisMaintaining the stemness of CSCs
Anchoring proteinsEzrinRegulation of actin polymerization by ROCK inhibitionMaintaining the properties of CSCs
MoesinEnhancement of positive feedback on the Wnt/β-catenin signaling pathway by increasing the expression of CD44Promoting CSC function
MerlinAdjusting the YAP/TAZ pathway via the merlin/NF2/YAP/TAZ axisPromoting CSC function
Capping and severing proteinsGelsolinDirect regulation of stem cell-associated transcription factorsMaintaining the properties of CSCs
CofilinActs by promoting EMT expressionMaintaining the properties of CSCs
Stabilizing proteinsTropomodulinIncreased expression of MMP-13 and NF-κB and the activation of the PI3K–AKT signaling pathwayPromoting CSC function
Signaling proteinsENA/VASPECM-mediated β1-integrin-FAK–YAP/TAZ signaling pathwayMaintaining the properties of CSCs
TubulinRegulating EMT and contributing to the formation of lamellar filopodiaPromoting CSC function
Microtubule lattice-binding proteinsTauActivating the MAPK pathway by binding to PI3KMaintaining the properties of CSCs
kataninActivation of JNK by cutting cell microtubules into short segmentsPromoting CSC function
ATIP3Inhibition through ATIP3/ERK1/2-Snai2 signalingInhibiting the properties of CSCs
Microtubule motor proteinsKinesinsPromoting the expression of stem transcription factors (NANOG and OCT4)Maintaining the properties of CSCs
Multiple site microtubule-binding proteinsSTMN1Activates Rho by promoting microtubule depolymerizationPromoting CSC function
Microtubule minus-end-binding proteinsCAMSAPsInhibition of Akt activity through microtubule regulationInhibiting the properties of CSCs
MyosinInvolved in promoting the EMT and enhancing the nuclear translocation of CSCMaintaining the properties of CSCs
Intermediate filamentsVimentinRegulation of EMT-related genes, including Twist, Snail, ZEB1/2 and SlugMaintaining the properties of CSCs
NestinUpregulation of VEGF, cancer stem cell markers, and proteins that activate Wnt/β-catenin to initiate proliferationPromoting CSC function
KeratinsInteracting with the β5-integrin/c-Met signaling pathwayPromoting CSC function
Binding to β4-integrin/FAK or Src or β-catenin
Table 2. Overview of clinical trials and experiments targeting the cytoskeleton.
Table 2. Overview of clinical trials and experiments targeting the cytoskeleton.
CategoriesDrug NameMechanismClinical TrialNCT Registry Number/Ref.
ActinSMIFH2Inhibiting actin nucleation and elongationExperimental[254]
ToradolGTPase inhibitionActive, not recruitingNCT02470299
MBQ-167Rac/Cdc42 inhibitorExperimental[256]
ProfilinC1 and C2Preventing profilin from interacting with actin monomersExperimental[267]
Tβ4Tβ4 inhibitorsSilencing the Tβ4 geneExperimental[172,268]
FascinNP-G2-044Inhibiting fascin-1-directed actin remodelingCompletedNCT03199586
Compounds 3 and 14Downregulating fascin-1Experimental[271]
RaltegravirInhibitor of human immunodeficiency virus 1 integraseExperimental[274]
SalinomycinFascin-1 inhibitionExperimental[275]
ImipramineFascin-1 inhibitionEarly Phase 1NCT03122444
IINCT04863950
EzrinNSC305787Inhibiting the phosphorylation of ezrinExperimental[278]
NSC668394Experimental[279]
Cytochalasin BInhibition of actin assemblyExperimental[281]
LY294002PI3K inhibitorExperimental[197]
MK2206AKT inhibitor
BAY43-9006Multi-kinase inhibitorExperimental[283]
BaicalinInhibitor of ezrinExperimental[284]
CelastrolImpairing the phosphorylation of ezrinExperimental[285]
MicrotubulesPaclitaxelStabilizing microtubulesClinical medication[287]
TaxanesInhibitors of microtubule dynamicsClinical medication[12]
Vinca alkaloids
VimentinWithaferin-AInducing vimentin cleavageExperimental[290]
KinesinPurine compound 5aRegulating katanin’s cut-off activitiesExperimental[223]
IspinesibA kinesin spindle protein inhibitorCompletedNCT00089973
SB-743921NCT00136513
StathminAnti-stathmin adenovirusCell cycle inhibitionExperimental[300]
Aurora A shRNAInhibitor of stathminExperimental[301]
MitochondriaβIIPKCInhibitor of mitochondrial fusion proteinsExperimental[303]
MetforminInhibitors of OXPHOSExperimental[304]
BenzforminExperimental[305]
IM156Metformin derivativeExperimental[306]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Li, Y.; Wang, D.; Ge, H.; Güngör, C.; Gong, X.; Chen, Y. Cytoskeletal and Cytoskeleton-Associated Proteins: Key Regulators of Cancer Stem Cell Properties. Pharmaceuticals 2022, 15, 1369. https://doi.org/10.3390/ph15111369

AMA Style

Li Y, Wang D, Ge H, Güngör C, Gong X, Chen Y. Cytoskeletal and Cytoskeleton-Associated Proteins: Key Regulators of Cancer Stem Cell Properties. Pharmaceuticals. 2022; 15(11):1369. https://doi.org/10.3390/ph15111369

Chicago/Turabian Style

Li, Yuqiang, Dan Wang, Heming Ge, Cenap Güngör, Xuejun Gong, and Yongheng Chen. 2022. "Cytoskeletal and Cytoskeleton-Associated Proteins: Key Regulators of Cancer Stem Cell Properties" Pharmaceuticals 15, no. 11: 1369. https://doi.org/10.3390/ph15111369

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop