Skip to main content

RhoA-ROCK2 signaling possesses complex pathophysiological functions in cancer progression and shows promising therapeutic potential

Abstract

The Rho GTPase signaling pathway is responsible for cell-specific processes, including actin cytoskeleton organization, cell motility, cell division, and the transcription of specific genes. The implications of RhoA and the downstream effector ROCK2 in cancer epithelial-mesenchymal transition, migration, invasion, and therapy resistance associated with stem cells highlight the potential of targeting RhoA/ROCK2 signaling in therapy. Tumor relapse can occur due to cancer cells that do not fully respond to adjuvant chemoradiotherapy, targeted therapy, or immunotherapy. Rho signaling-mediated mitotic defects and cytokinesis failure lead to asymmetric cell division, allowing cells to form polyploids to escape cytotoxicity and promote tumor recurrence and metastasis. In this review, we elucidate the significance of RhoA/ROCK2 in the mechanisms of cancer progression and summarize their inhibitors that may improve treatment strategies.

Introduction

Rho GTPases are the members of the Ras superfamily and are known for their important roles in regulating the actin cytoskeleton. Rho, Rac, and Cdc42 are the most extensively characterized Rho GTPases. The Rho subfamily contains the three isoforms RhoA, RhoB, and RhoC [1]. The Rho family proteins have lipid modifications that allow them to localize to cell membranes and act as molecular switches, cycling between active GTP-bound and inactive GDP-bound forms. Binding to GTP is promoted by Rho guanine nucleotide exchange factors (Rho GEFs), and they can interact with effectors or target molecules in the GTP-bound form to initiate downstream responses, whereas GTP hydrolysis is catalyzed by Rho GTPase-activating proteins (Rho GAPs), reverting the proteins to the GDP-bound state to complete the cycle and terminate signaling. Rho GDP-dissociation inhibitors (GDIs) sequester GDP-bound Rho GTPases in the cytoplasm and inhibit GDP and GTP exchange activity [1,2,3].

Rho proteins play critical roles in actin cytoskeleton remodeling, influencing cell motility and supporting cellular processes such as migration, invasion, and tube formation. RhoA is involved in gene transcription, cell cycle progression, and cell transformation and RhoA plays a crucial role in the progression and development of many malignancies, such as breast, gastric, and colorectal cancer [4]. It regulates cancer cell survival, proliferation, adhesion, the cell cycle, and gene transcription [5, 6]. Mutations in RhoA have been reported in many human cancers, wherein they contribute to the malignant processes [7]. RhoA is overexpressed in many tumors and has been associated with poor prognosis [8], and its inhibition results in decreased tumor proliferation and migration [9]. Upregulated RhoC also regulates several tumor phenotypes [10, 11] and induces TGFβ signaling mediated epithelial-mesenchymal transition (EMT) in ovarian and cervical cancers [12, 13]. In contrast, RhoB has the opposite function, acting as a negative modifier or suppressor gene in cancer cells. In a variety of solid tumors, such as lung, head and neck, and brain cancers, RhoB levels decrease as tumor progresses [14].

Activated RhoA triggers one of its downstream effectors Rho-associated coiled-coil-containing protein kinases, ROCK1 or ROCK2. These kinases play a vital role in stress fiber and focal adhesion formation, smooth muscle contraction, neurite retraction, microvilli formation, and cell migration. ROCK2 also plays an important role in tumor progression. In this review, we discuss the role of RhoA/ROCK2 signaling in normal cells and its correlation with cancer cell invasion, migration, division, tumor stemness, and resistance to therapy. This study aimed to understand tumor progression and identify potential targeting strategies.

Structure of Rho proteins

The gene encoding RhoA (former gene name: aplysia Ras-related homolog, ARH12) is located on chromosome 3 in humas, and its precursor mRNA is subject to highly variable alternative splicing, resulting in seven known transcript variants for RhoA, which are then translated into six different RhoA isoforms [15]. The protein structures of Rho subfamily members have been intensively studied for decades, revealing their binding to different nucleotides or nucleotide analogs at different activity states and their interactions with different effectors, including GEFs, GAPs, and GDIs.

RhoA, RhoB, and RhoC possess a relatively conserved N-terminal portion containing a G domain (also known as RhoA-like domain) and a less-conserved C-terminal tail. The conformational changes from activated to inactivated states are mainly restricted to two surface loop regions: switch I and switch II [16]. The G domain consists of five conserved sequence motifs (G1 to G5) that are involved in nucleotide binding and hydrolysis [17]. The conformational changes in switches I (G2) and II (G3) from the inactive GDP-bound to the active GTP-bound state are also a prerequisite for the GTPase to bind to effector proteins. This possibly occurs via interactions with hydrophobic residues in switch regions that are exposed in the active state of the GTPase [18]. In addition to the switch regions, an inserted α-helix (located between the G4 and G5 motifs) has been shown to be important for effector protein interaction [19]. The crystal structures of the GTPase-binding domains (GBDs) of the RhoA-associated coiled-coil kinase (ROCK)-RhoA complexes revealed that, as predicted from their primary structure, these domains formed α-helical coiled coils arranged in a parallel fashion. A 13-residue left-handed coiled-coil in the C-terminal portion of the ROCK-GBD binds exclusively to the switch and α2 regions of RhoA and is considered the minimal sequence required for Rho-interacting motif activity [20].

The C-terminal hypervariable region terminates with a common sequence known as CAAX (where C represents cysteine, A is an aliphatic amino acid, and X is any amino acid), and post-translational modifications in this sequence, including prenylation, endoproteolysis, and carboxyl methylation, are critical for the subcellular localization of Rho GTPases [21] (Fig. 1). The X residue determines which isoprenoid will be added to the cysteine. If the amino acid in the X position is leucine, isoleucine, or phenylalanine, as in the Rho/Rac family of proteins, geranylgeranyltransferase (GGT) modifies CAAX Cys residues, while farnesyltransferase (FT) utilizes mostly serine, methionine, or as other amino acids in the X position [22, 23]. Prenylation is followed by truncation and methylation and the lipid moiety makes the C-terminus hydrophobic and helps anchor it to membranes [24]. In contrast to RhoA and RhoC proteins, both farnesylated and geranylgeranylated forms of RhoB (RhoB-F and RhoB-GG) are present in COS and Rat1/ ras cells [25,26,27].

Fig. 1
figure 1

Sequential schematic of the protein structural elements and binding sites of RhoA as retrieved from the PDB database (ID number: 1s1c). Different colors of a single letter represent different post-translational modification sites: red for phosphoserine, green for glycosylation sites, and blue for ubiquitination sites. The red-shaded background on the sequence represents the site that binds to GTP, and the blue-shaded background is the CAAX sequence that GDI interacts with. The purple curve line and red boxes below the sequence correspond to α-helices and β-strands, respectively

Post-translational modifications of RhoA

To ensure appropriate spatiotemporal activation, the GTPases undergo post-translational modifications. The most well-studied are the phosphorylation of serine, threonine, and tyrosine residues by several critical kinases [28]. RhoA is phosphorylated by cAMP-dependent protein kinase (PKA), cGMP-dependent protein kinase (PKG), and protein kinase C (PKC) on Ser 188 [29], whereas ste20-related kinase (SKL) and AMP-activated protein kinase subunit alpha 1 (AMPKα1) inactivate Ser188 [30, 31]. Phosphorylation of Ser188 deactivates RhoA by increasing its interaction with Rho GDI, leading to its translocation from the site of action at the membrane to the cytoplasm [32] and causing the collapse of actin stress fibers. Extracellular signal-regulated kinase (ERK) phosphorylates RhoA at Ser88 and Thr100 to upregulate RhoA [33]. Mammalian Ste20-like kinase 3 (Mst3) phosphorylates RhoA on Ser26 to inactivate RhoA [34]. In addition, other tyrosine residues, Tyr34, Tyr66, and Tyr42, are regulated by different tyrosine kinases such as Bcr-Abl, Src, and c-Met [35,36,37]. Additionally, RhoA undergoes ubiquitination, leading to various outcomes, such as re-localization or degradation [38]. RhoA is ubiquitylated by E3 ubiquitin ligase complexes, such as SMAD-specific E3 ubiquitin protein ligase (SMURF1), Cullin3, and RNF8, leading to proteasomal degradation and regulation of protein turnover [39,40,41,42]. For RhoA, the prerequisite process involves the addition of a geranylgeranyl moiety to Cys190 in the CAAX motif, followed by proteolysis and methylation [21, 43].

Role of RhoA in cytoskeletal rearrangement

Cytoskeletal rearrangements play critical roles in the regulation of various cellular processes linked to transformation, including proliferation, contact inhibition and apoptosis [44]. When bound to GTP, RhoA interacts with many downstream effectors. ROCK and mammalian diaphanous-related protein (mDia) initiate a network of cytoplasmic and nuclear signaling cascades. mDia facilitates actin nucleation and polymerization and induces long, straight actin filaments [45, 46], and stabilizes and aligns microtubules in interphase cells [47]. Rho acts on ROCK to phosphorylate myosin phosphatase and myosin light chain kinase at Ser19 [48, 49], leading to actin filament formation and increased actin-myosin interactions [50, 51] and contribution to the invasive phenotype [52]. ROCK phosphorylates LIM kinase 1 (LIMK1) at Thr508 [53], then phosphorylates and inactivates cofilin (filament severing and depolymerization from the pointed end) to regulate actin filament stabilization, focal adhesion, and actin network assembly [54, 55].

RhoA-ROCK signaling is involved in the cell cycle

Rho proteins regulate cell cycle proteins that are mainly involved in the G1/S transition [56, 57]. Activation of RhoA and Rho-dependent stress fiber accumulation upregulates cyclin D1 expression levels and consequent G1-phase progression. Macrophage migration inhibitory factor promotes the activation of the canonical extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase cascade and cyclin D1 expression by stimulating Rho GTPase activity and downstream signaling to stress fiber formation and subsequent progression through G1 to S phase [58,59,60,61]. In addition, RhoA increases the ubiquitination-dependent degradation of p27 (cyclin-dependent kinase inhibitor p27kip1) by contributing to an earlier stage of G1 progression [62]. Inhibition of RhoA and ROCK resulted in increased p21 (cell cycle inhibitor p21Cip1/Waf1) expression, which is dependent on phosphorylated ERK, resulting in decreased cell proliferation [63, 64].

RhoA regulates mitosis

Mitotic progression is highly regulated and involves the dynamic modulation of cell shape and morphology, primarily through the remodeling of the actin and microtubule cytoskeleton. Rho GTPases in animal somatic cells regulate the mitotic stages from prophase to telophase, and then to cytokinesis [57, 65,66,67]. During prometaphase, chromosomes are captured at kinetochores as mitotic spindles assemble. The presence of all chromosomes at the spindle equator is necessary for accurate chromosome segregation; however, this process is prone to error [68].

Cytokinesis is a finely orchestrated process that requires the formation and progression of a cleavage furrow generated by the contraction of an actomyosin contractile ring anchored to the plasma membrane by cytoskeletal proteins [69]. At the onset of mitosis, actin cytoskeleton rearrangement, de-adhesion, and an increase in cortical rigidity accompany mitotic cell rounding, during which a flat cell becomes spherical [70]; Cdk1/cyclin B1 mediates the phosphorylation of RhoA regulators causing a global increase in RhoA activity; activated RhoA acts on ROCK to alter cell shape [71]. This is followed by nuclear envelope rupture during mitosis and an increase in RhoA activity and the concentration of active RhoA in the cell cortex. Following chromosome segregation, cells immediately undergo highly ordered cytoplasmic division, and RhoA plays a central role in regulating cytokinesis [72]. RhoA activation is required to stabilize midzone microtubules and maintain midzone structures after anaphase onset or during cytokinesis [73]. Recruitment of RhoA determines the cleavage sites and promotes contractile ring assembly and furrow ingression [74, 75] or induces polyploidy if actin filaments are not concentrated at furrow sites [76, 77]. During cleavage, daughter cells move apart to expose the intercellular bridges that stretch between them. Cytokinesis is completed when the intracellular bridge is severed.

Intermediate filament (IF) proteins are located in the cleavage furrow and must be phosphorylated by ROCK during late cytokinesis, which locally breaks down IFs and separates IF networks efficiently [78, 79]. If cytoplasmic division fails and intermediates are formed in this state, nascent daughter cells may remain attached. This results in the generation of binucleated aneuploid or polyploid giant cells that may lead to genomic instability and contribute to the initiation and/or progression of tumorigenesis. Megakaryocytes (MKs), which are naturally polyploid cells that give rise to platelets, require RhoA/ROCK/F-actin for cytoplasmic maturation, and inhibition of this pathway leads to macrothrombocytopenia [80,81,82].

RhoA-ROCK2 pathway and asymmetric cell division (ACD)

The RhoA-ROCK2 signaling pathway plays a crucial role in cytoskeletal rearrangement and regulation during somatic cell division. Deviations in this pathway can result in aneuploidy formation or ACD, which can damage various cellular processes such as plasma membrane invagination, cleavage site positioning or specification, cleavage furrow formation, ingression, midbody formation, and cell separation [65, 69, 83]. Aneuploidy is a common characteristic of cancer cells present in approximately 90% of solid human tumors and 50% of hematopoietic cancers. Hypoxia stimulates the NPY/Y5R axis and leads to RhoA overactivation, cytokinesis defects, and polyploidy, triggering chromosomal instability and bone metastasis in Ewing sarcoma [84] (Fig. 2).

Fig. 2
figure 2

RhoA signaling pathway regulates cell asymmetric division. (a) Chromosomes are arranged on the equatorial plate with two levels of centromeres pulling them laterally. (b) The central spindle recruits the Rho GEF and delivers it to the plasma membrane where it activates RhoA. Activated RhoA directs the assembly of the contractile ring containing the filamentous actin myosin II and contracts to change cell shape. (c) Failure of cell division owing to lack of contractility or centrosome can lead to asymmetric cell division

Dynamic contractile ring control mitosis through RhoA recruitment effectors

The contractile forces required for furrow ingression are provided by a ring of filamentous actin and myosin II, which are juxtaposed with the cell membrane at the equator of the dividing cells. RhoA-dependent ROCK activity is required for myosin II recruitment to the cortex, whereas myosin light chain kinase (MLCK) activity promotes myosin II turnover [85]. Uniform tension distribution around the ring depends on continuous turnover of actin filaments; however, a nonuniform tension distribution around the ring leads to peeling off of actin and myosin [86]. Cofilin is activated by myosin II to polymerize actin and assemble the contractile ring [87], rapidly accumulated during the late stages of furrowing, and eventually enriched at the midbody [88]. Cofilin, an actin-binding protein, contributes to turnover by severing actin filaments for normal function. The absence of cofilin results in longer filaments, higher ring tension, and increased actin dissociation from myosin in fission yeast [89].

Furthermore, F-actin is also required for anchoring the mitotic spindle to the cell cortex and determining the direction of spindle movement [90]. It is plausible that in the absence of cofilin, the excessive amount of actin forms filaments during mitosis that prevent proper spindle positioning and manifest as chromosome segregation defects and aberrant cytokinesis [91]. After silencing endogenous cofilin, cells lacking nuclear localization fail to sever nuclear F-actin during mitotic exit, resulting in defective nuclear volume expansion and chromatin decomposition [92].

Rho GEFs and GAPs involved in ACD through act on RhoA

Rho GAPs such as IQGAP [93,94,95], ARHGAP19 [96, 97], and ARHGAP11A [98], affect the recruitment and localization of key cytokinesis, or involved in spindle orientation. MgcRacGAP (RACGAP1, named Cyk-4 in C. elegans, RacGAP50C in Drosophila [99, 100]) localizes to the central spindle and contractile ring and binds the kinesin-like protein MKLP-1 to form an evolutionarily conserved complex called centralspindlin. It is essential not only for cytoplasmic division of somatic cells but also for cytoplasmic contraction, leading to intercellular bridges [101]. Rho GEF Ect2 localization to the central spindle depends on centralspindlin [102]. Ect2 depletion impairs microtubule attachment to kinetochores and causes prometaphase delay and abnormal chromosome segregation [103]. Rho GEF-H1/Lfc associates with mitotic spindles and spindle microtubules. Disruption of Lfc or the central spindle results in a reduction of active RhoA concentration in the equatorial plane, which in turn leads to a delay in mitosis [104,105,106]. MP-GAP (Rho GAP) was shown to target RhoA during mitosis/cytokinesis, and depletion caused excessive RhoA activation in M-phase, leading to the uncontrolled formation of large cortical protrusions and late cytokinesis failure [107].

Proper completion of cytokinesis requires the interplay of Rho GEFs and GAPs to appropriately regulate RhoA levels. The GRAF (Rho GAP) and RhoGEF2 are in balance for proper activation of actomyosin ring contraction [108]. Endogenous p190RhoGAP colocalize with Ect2 at the cleavage furrow during cell division [109]. Cells with ectopic or loss of expression of the protein have reduced Rho GTP levels at the cleavage furrow and become multinucleated or fail to divide [110].

RhoA-ROCK signaling involved in ACD through centrosome disorganization

The centrosome is a small, self-replicating organelle that coordinates with mitosis [111, 112]. Abnormalities in centriole duplication, disengagement, or loss are hallmarks of cancer [113, 114], these abnormalities cause delays in the bipolar spindle assembly, high rates of chromosomal instability, and aneuploidy in vertebrate somatic cells [115, 116]. Signaling components, such as kinases and phosphatases, including ROCK, are associated with centrosomes, spindle poles, and microtubule organizing centers (MTOCs) [117]. They could maintain centrosome stable and separation. ROCK2, associated with nucleophosmin (NPM), prevents aberrant centrosome amplification and a high frequency of multinucleated cells [118, 119]. Inhibition of ROCK1/2 affects chromosome segregation, bypasses the spindle assembly checkpoint (SAC), blocks late cytokinesis, induces microtubule-dependent centrosome fragmentation, and increases the distance between mother and daughter centrioles in G1 cells [120].

RhoA indirectly modifies the actin cytoskeleton through ROCK1, but not ROCK2, and directly affects centriole structure and function through centriole-associated factors [121]. Similarly to ROCK, downstream LIMK inhibition also induces centrosome fragmentation [122]. Myosin II is also essential for centrosome separation and positioning during mitotic spindle assembly [123]. In addition, during interphase and mitosis, p190B Rho GAP and MgcRacGAP localize to chromosomal centromeres and provides epigenetic centromere maintenance [124, 125]. The Rho GEF ARHGEF10 regulates RhoA and controls centrosome duplication through its binding partner, the motor protein KIF3B, which colocalizes with ARHGEF10 at the centrosome [126].

Cofilin, downstream of RhoA, regulates ACD during meiosis

Meiosis occurs in oocytes that lack centrosomes [127] and possess a dynamic cytoplasmic actin network. This network is critical for spindle migration during meiosis and functions in cooperation with various actin-binding proteins [128]. Cofilin is primarily distributed around spindles. LIMK overexpression, which interferes with cofilin activation during maturation, results in the depletion of cytoplasmic F-actin. This depletion leads to defects in spindle migration and polar body extrusion [129]. Cyclase-associated proteins (CAPs) can promote actin disassembly by enhancing the actin-severing activity of cofilin. The knockdown of CAP1 leads to the accumulation of excessive actin filaments near the spindles, which impairs meiotic spindle migration and asymmetric division [130].

RhoA regulates the migration of non-tumor cells

The actin cytoskeleton endows the cell with shape, structure, and polarity and undergoes constant remodeling. Lamellipodia, filopodia, stress fibers, and focal adhesions constitute the structural framework of the actin cytoskeleton [131]. In normal epithelia, RhoA helps generate epithelial polarity and junction assembly and function [132]. The role of RhoA is complex, as the production of actin assembly drives protrusions at the front, coordinating actomyosin contractility-driven retraction at the tail to facilitate movement during cell migration in 2-D environments [133]. For example, in migrating leukocytes, cell body and tail contraction depend on actomyosin contractility and can be regulated by RhoA/ROCK [134, 135]. However, excessive activation of RhoA causes an increase in the contractile force that pulls endothelial cells and disrupts barrier function. Cofilin is required for and promotes lamellipodium extension and cell migration [136]. It has also been reported that RhoA cooperates with Rac1 and Cdc42 to induce membrane ruffles through the recruitment of mDia, implying amoeboid-like motility [137]. These findings are of particular interest with respect to explaining of epithelial cell migration.

Cell adhesion activates RhoA, leading to the activation of ROCK and mDia, organization of actomyosin bundles into stress fibers, and the formation of focal adhesion complexes composed of vinculin, paxilin, and focal adhesion kinase [51]. Tyrosine kinases are essential in signal transduction from integrins to RhoA; tyrosine phosphorylation of the focal adhesion plaque alters the assembly of actin fibers by RhoA/ROCK, which then effect the formation and aggregation of focal adhesions [138, 139]. Focal complex confined to lamellipodia can be induced by high level of Rac and low level of Rho, enlarge and elongate centripetally into typical focal contacts upon upregulation of Rho, and turn over for cells to migrate [140].

RhoA-ROCK signaling pathway promotes cancer progression

Rho GTPases, in addition to many other cellular functions, are components of the signaling network that coordinates cell proliferation with microenvironmental dynamics, and their deregulation contributes to malignant transformation and cancer. In addition to changing the adhesive repertoire, cancer cells employ developmental processes to gain migratory and invasive properties that involve dramatic reorganization of the actin cytoskeleton and concomitant formation of membrane protrusions, which are required for the invasive growth of cancer cells (Fig. 3).

Fig. 3
figure 3

RhoA is overexpressed in cancer and promotes cancer progression via RhoA/ROCK2 signaling pathway. RhoA and its downstream effectors Rock2, myosin II, LIMK, and cofilin play critical roles during G1-S phase progression by regulating the expression of cell cycle proteins p21 and cyclin D1. RhoA regulated by Rho GEFs and GAPs can recruit effectors to form a contraction ring during cell cytokinesis, and the loss of contractile forces can cause asymmetric division. RhoA-mediated actin assembly, protrusion at the front of the cell, and contraction at the body of the cell enhance motility and promote cancer cell invasion and migration. In addtion, RhoA plays an important role in epithelial-mesenchymal transition and the formation of vasculogenic mimicry network. These processes drive cancer stem cells to tumor progression

RhoA promotes the migration and invasion of cancer cells by regulating EMT

During EMT, in which epithelial cells gradually dedifferentiate, polarized epithelial cells lose their adhesive properties and acquire a mesenchymal cell phenotype. As a result, the cells acquire migratory and invasive properties that allow them to penetrate the extracellular matrix (ECM) more easily [141, 142]. Cell movement in the ECM is a multistep process that requires reorganization of the actin cytoskeleton and formation of membrane protrusions. Increased intracellular pressure via RhoA-ROCK signaling leads to blebbing, thus causing amoeboid migration, while regulating tail retraction during mesenchymal migration [143, 144]. Apart from lamellipodia and filopodia, tumor cells also form lobopodia, which are blunt cylindrical protrusions formed by intracellular pressure and have been discovered in the three-dimensional extracellular matrix [145].

In addition, cells also form invadopodia and podosomes, actin-rich structures that proteolytically degrade the ECM for 3D migration [146]. Proteases are released to remodel the extracellular matrix by proteolysis, such as the levels of Matrix metalloproteinase-9 correlated with invasive ability [147].

RhoA enhances the formation of vasculogenic mimicry in malignant tumors

In addition to tumor angiogenesis, aggressive tumors acquire microcirculation through vasculogenic mimicry (VM), which tumor cells form by acquiring plasticity to mimic embryonic vasculogenic networks and connect with those of the endothelium for invasion and metastasis. It is known that, RhoA/ ROCK plays an important mediated role in the process of cancer cell VM formation [148]. RhoA signaling is involved in the maintenance of actin organization, induction of migration, and tube formation in the VM [149]. Hypoxia enhances RhoA/ROCK 2 and Rac1/Pak activity, stabilizes hypoxia-inducible factor-1α (HIF-1α) protein, and induces VM formation through HIF-1α stabilization and EMT with vimentin phosphate activation [150]. In addition, axon guidance factor Sema4D activates RhoA/ROCK pathway and regulates tumor cell plasticity and migration to achieve VM in non-small cell lung cancer [151]. ROCK mediates TGF-β1-induced EMT and cancer stemness activity to participate in VM formation [152]. The traditional herbal medicines Baicalein [153] and Incarvine C [154] suppress VM by inhibiting the RhoA/ROCK signaling pathway.

RhoA signaling pathway promotes the stemness of cancer cell

Rho GTPases enhance the stemness of cancer cells by interacting with other signaling pathways, such as Notch and Wnt [155,156,157]. Wnt3A was found to promote superoxide generation by activating the phosphorylation of RhoA, in addition to Wnt3/RhoA/ROCK signaling pathway being involved in adhesion-mediated drug resistance of multiple myeloma [36, 158]. Loss of p130 induced expression of Wnt5A, which selectively increased RhoA transcription and drove tumorigenesis in small cell lung cancer cells [159].

YAP/TAZ are sensors of the structural and mechanical features of the cellular microenvironment. Their activation induces CSC characteristics, such as proliferation, chemoresistance, and metastasis [160]. RhoA mediates YAP1 dephosphorylation and transport to the nucleus, inducing autophagy and promoting the migration of liver CSCs [161]. Protease-activated receptor 1 (PAR1) activation inhibits the Lats kinase associated with the Hippo-YAP pathway via Rho. Inhibition of Lats kinase results in increased nuclear localization of dephosphorylated YAP, leading to EMT [162]. Another study has shown that inhibition of the RhoA GTPase/F-actin pathway may be a useful approach for suppressing YAP/TAZ activity and limit breast CSC formation [163]. In cancer-associated fibroblasts, RhoA knockout can reduce branching and meshwork formation and may promote CSC-like properties [164].

RhoA signaling pathway is associated with the formation of polyploid giant cancer cells

Tumors are composed of non-homogeneous cell populations that exhibit varying degrees of genetic and functional heterogeneity [165]. Polyploid cancer cells (PGCCs) are a subtype of CSCs that express stem cell properties and have differentiation potential. PGCCs are large and contain single giant nuclei or multiple nuclei with significant variations in shape, chromatin pattern, and number of nucleoli; they display an ACD by budding or bursting [166,167,168]. PGCCs confer resistance to DNA damage and contribute to the formation of complex tumor cell karyotypes by promoting aneuploidy and chromosomal instability [169,170,171]. The PGCCs exhibited significant differences in the organization of actin stress fibers, including the presence of longer and thicker stress fiber bundles. Increased cytoskeletal rigidity and nuclear structure are largely regulated by the RhoA-ROCK1 signaling pathway and actin cytoskeletal dynamics, which are essential for protecting cells from injury [172]. Therefore, the regulation of the RhoA/ROCK pathway is critical to understanding how PGCCs promote tumor progression.

RhoA-ROCK2 signaling is involved in chemoradiation therapy resistance

Although most cancer cells tend to become addicted to therapy, PGCCs or other CSCs show low sensitivity, undergo transient dormancy, and can evade treatment and cause cancer relapse after emerging from their dormant state [173, 174]. For example, RhoA signaling promoted CSC phenotypes in diffuse-type gastric adenocarcinoma cells, and RhoA inhibition could reverse chemotherapy resistance both in vivo and in vitro [175]. The mechanisms of chemoresistance in cancer cells are complicated, and the main mechanisms include increased drug efflux mediated by membrane transport proteins [176,177,178], reduction of Bcl-2 family mediated cell apoptosis [179, 180], and reduction of topoisomerase activity, leading to decreased affinity of topoisomerase to its target cells [181]. RhoA is closely associated with multidrug resistance-associated proteins (MRPs) in the development of chemoresistance.

RhoA was mediated by GPR56 and enhanced drug resistance through upregulation of MDR1 levels [182]. RhoA knockdown increases NF-κB activation, which induces nitric oxide production and leads to tyrosine nitration of multidrug resistance protein 3 (MRP3), thereby reducing doxorubicin efflux and reversing chemoresistance [183]. Besides, inhibition of RhoA rescued the resistance to CPT-11 by inhibiting MRP1 and GSTP1 expression and promoting apoptosis [184]. Notably, RhoA exerted different effects on chemoresistance to different drugs. For example, downregulation of RhoA decreased the chemoresistance of cells to doxorubicin due to MRP1 internalization and increased doxorubicin accumulation but decreased chemoresistance to cisplatin due to decreased cisplatin influx [185, 186]. In osteosarcoma, RhoA enhances therapy resistance and suppresses apoptosis after photodynamic treatment through the Hippo/YAP pathway [187]. Mechanically, RhoA knockdown alleviates the antagonism of HIF-1α to hypoxia-induced apoptosis [188]. There is also evidence that RhoA facilitates brain metastasis during the evolution of Osimertinib-resistance in NSCLC cells [189].

In addition, elevated ROCK2 expression is associated with chemoresistance. Inhibition of ROCK2 signaling sensitizes drug-resistant prostate cancer to enzalutamide [190] and gemcitabine [191]. In MGMT-low (O6-methylguanine-DNA methyltransferase) temozolomide (TMZ) resistant glioma cells, overactive ROCK2 increased homologous recombination repair and decreased TMZ sensitivity [192]. Both sunitinib and everolimus treatment significantly enhanced ROCK2 phosphorylation and subsequent β-catenin nuclear translocation [193]. The decreased expression of ARHGAP18 (a Rho GAP protein) leads to the increase of RhoA/ ROCK1 signaling pathway and promotes glucocorticoid resistance by antiapoptotic of leukemia cells [194].

There are reports that implicating the RhoA/ROCK pathway in radiation resistance [195]. Radioresistance in glioma cells could be induced by activation of the DNA repair pathway and increased levels of reactive oxygen species (ROS), resulting in the enrichment of CD133 + CSCs [196, 197]. On the one hand, modulation of RhoA activity sensitized cells to γ-irradiation by attenuating the DNA damage response and repair pathways [198]. In addition, activated RhoA is found in the nucleus of tumor cells after irradiation and regulates resistance by modulating survivin activity [199, 200]. In contrast, RhoA is a downstream target of heterogeneous nuclear ribonucleoprotein C1/C2, and RhoA inhibition can hinder the activity of cancer-associated fibroblasts and weaken the radiation resistance of pancreatic tumors [201]. ROCK inhibitor reduced survival and DNA repair capacity in wild-type p53 cells by inhibiting NHEJ and NER pathways with reduction of γH2AX foci and accumulation of strand breaks [202].

Small molecular inhibitors targeting RhoA-ROCK2 signaling

The implications of Rho GTPases and their upstream regulators or downstream effectors in the transformation, migration, invasion, and tumorigenesis of various CSCs highlight the potential of Rho GTPase targeting in cancer therapy [203]. First, targeting RhoA inhibitors such as Botulinum C3 exoenzyme, Grincamycin B, and Riboprine (N6-isopentenyladenosine) inhibits RhoA activity and attenuates stemness, inhibits the VM network formation, and suppresses tumor growth and invasion [204, 205]. Inhibitors of the RhoA/ROCK2 pathway shown in Table 1. The RhoA inhibitor Rhosin, which specifically binds to RhoA, was the first RhoA subfamily specific inhibitor developed to target GEF activation, and has been used in several cancer cells [139, 206, 218]. CCG-1423 is an inhibitor of RhoA/C that disrupts the transcriptional response of the Rho pathway [207]. CCG-100,602 and CCG-203,971 are new second-generation RhoA inhibitors with lower toxicity and higher selectivity and potency than those of CCG-1423 [208]. JK-136 and JK-139, two of the anti-RhoA hydrazide derivatives, were found to inhibit gastric cancer in mice [209].

Table 1 Inhibitors of the RhoA/ROCK2 pathway

The implications of Rho GTPases and their upstream regulators or downstream effectors in the transformation, migration, invasion, and tumorigenesis of various CSCs highlight the potential of Rho GTPase targeting in cancer therapy [203]. First, targeting RhoA inhibitors such as Botulinum C3 exoenzyme, Grincamycin B, and Riboprine (N6-isopentenyladenosine) inhibits RhoA activity and attenuates stemness, inhibits the VM network formation, and suppresses tumor growth and invasion [204, 205]. Inhibitors of the RhoA/ROCK2 pathway shown in Table 1. The RhoA inhibitor Rhosin, which specifically binds to RhoA, was the first RhoA subfamily specific inhibitor developed to target GEF activation, and has been used in several cancer cells [206, 218, 139]. CCG-1423 is an inhibitor of RhoA/C that disrupts the transcriptional response of the Rho pathway [207]. CCG-100,602 and CCG-203,971 are new second-generation RhoA inhibitors with lower toxicity and higher selectivity and potency than those of CCG-1423 [208]. JK-136 and JK-139, two of the anti-RhoA hydrazide derivatives, were found to inhibit gastric cancer in mice [209].

ARHGEF12, also known as LARG, regulates RhoA activity and could regulate cell morphology and invasion [219], as well as the mechanical response to integrins, mesenchymal stem cell stemness [210, 220] Y16, a small-molecule inhibitor of ARHGEF12, has been reported to inhibit RhoA activity and suppress sphere formation in breast cancer cells [218, 221]. The activity of Rho GEF Vav was important for the development of head and neck squamous cell carcinoma and showed effective inhibition in vivo [222, 223].

Post-translational modifications of RhoA may be promising cellular targets for anticancer therapy. Lovastatin suppressed EGF-induced thyroid cancer cell invasiveness by reducing Rho geranylgeranylation, which in turn suppressed membrane translocation and the subsequent suppression of Rho/ROCK and FAK/paxillin signaling [224]. PTX-100 is an inhibitor of geranylgeranyl transferase-1 (GGT-1), which mediates RhoA prenylation. It has shown significant antitumor activity in mouse models of breast cancer and is well tolerated. Patients in their advanced stages of disease are currently being recruited for a phase 1 clinical trial (NCT03900442) [225, 226].

Subsequently, common ROCK chemical inhibitors include fasudil (HA-1077) and Y-27,632, which can decrease tumor growth, invasion, and metastasis [211, 227,228,229] and sensitize CSCs to chemotherapy and radiation [230]. Y-27,632 can reduce the contractility and collagen degradation capacity of both CSCs and non-CSCs by blocking microtubule and F-actin assembly [212]. In addition, Y-27,632 activated dormant breast cancer cells and disrupted cell junctions, thereby promoting cell proliferation, migration, and invasion [231]. Exposure of circulating breast cancer cells to Y-27,632 destabilizes the actin cortex and increases the formation of microtubules, which are microtubule-based structures that enhance their ability to reattach to the vasculature [232]. However, interestingly, transient and continuous supplementation of non-toxic concentrations of Y-27,632 and fasudil inhibits apoptosis, enhances the ability of cells to form spheres, and increases stem cell marker expression in glioblastoma stem cells [233]; Y-27,632 primes the transition of CD44−/low cells to generate CD44high cells, helping to maintain a CD44high fraction and tumorigenic diversity in colon cancer [234]. Other selective inhibitors of ROCK include PT262, RKI-1447, CCT129253, and AT13148; however, their evaluation as anti-metastatic agents have been restricted to solid tumor cells, such as breast cancer and melanoma [49, 213,214,215, 235]. Photodynamic therapy combined with a ROCK inhibitor (ripasudil) and an anti-PD-L1 antibody could reduce uveal melanoma and prevent metastasis [216]. Furthermore, HSD1590, a new compound inhibited the migration of breast cancer cell [217].

Conclusion

In summary, the tumorigenic potential of Rho GTPases has led to continuous investigation of their functions. Many hallmarks of cancer, including unlimited replicative potential, evasion of apoptosis, tissue invasion, and metastasis, may be related to abnormal cytoskeletal or matrix mechanics. Undoubtedly, the RhoA/ROCK2 signaling pathway regulates cancer stemness in response to chemotherapy and radiation therapy and is involved in aneuploidy formation through ACD. Most RhoA/ROCK inhibitors remain in laboratory use; nevertheless, their pharmaceutical development and research continue to hold promise. These inhibitors show promise as emerging targets to combat cancer progression and suppress tumor stemness.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

Rho GEFs:

Rho guanine nucleotide exchange factors

Rho GAPs:

Rho GTPase-activating proteins

Rho GDIs:

Rho GDP-dissociation inhibitor

ROCK:

Rho-associated coiled-coil-containing protein kinase

MLCK:

myosin light chain kinase

LIMK:

LIM kinase

PKA:

protein kinase A

MRLC:

myosin regulatory light chain

EMT:

epithelial-mesenchymal transition

ECM:

extracellular matrix

VM:

vasculogenic mimicry

ACD:

asymmetric cell division

MTOC:

microtubule organizing center

SAC:

spindle assembly checkpoint

PCM:

pericentriolar material

PGCCs:

Polyploid giant cancer cells

CSC:

cancer stem cell

MRPs:

resistance-associated proteins

TMZ:

temozolomide

References

  1. Hall A. Rho family GTPases. Biochem Soc Trans. 2012;40(6):1378–82.

    Article  CAS  PubMed  Google Scholar 

  2. Bishop AL, Hall A. Rho GTPases and their effector proteins. Biochem J. 2000;348(Pt 2):241–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Schwartz M. Rho signalling at a glance. J Cell Sci. 2004;117(Pt 23):5457–8.

    Article  CAS  PubMed  Google Scholar 

  4. Sahai E, Marshall CJ. RHO-GTPases and cancer. Nat Rev Cancer. 2002;2(2):133–42.

    Article  PubMed  Google Scholar 

  5. Svensmark JH, Brakebusch C. Rho GTPases in cancer: friend or foe? Oncogene. 2019;38(50):7447–56.

    Article  CAS  PubMed  Google Scholar 

  6. Haga RB, Ridley AJ. Rho GTPases: regulation and roles in cancer cell biology. Small GTPases. 2016;7(4):207–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kataoka K, Ogawa S. Variegated RHOA mutations in human cancers. Exp Hematol. 2016;44(12):1123–9.

    Article  CAS  PubMed  Google Scholar 

  8. Chen W, Delongchamps NB, Mao K, Beuvon F, Peyromaure M, Liu Z, Dinh-Xuan AT. High RhoA expression at the tumor front in clinically localized prostate cancer and association with poor tumor differentiation. Oncol Lett. 2016;11(2):1375–81.

    Article  CAS  PubMed  Google Scholar 

  9. Jeong D, Park S, Kim H, Kim CJ, Ahn TS, Bae SB, Kim HJ, Kim TH, Im J, Lee MS, et al. RhoA is associated with invasion and poor prognosis in colorectal cancer. Int J Oncol. 2016;48(2):714–22.

    Article  CAS  PubMed  Google Scholar 

  10. Xie S, Zhu M, Lv G, Geng Y, Chen G, Ma J, Wang G. Overexpression of Ras homologous C (RhoC) induces malignant transformation of hepatocytes in vitro and in nude mouse xenografts. PLoS ONE. 2013;8(1):e54493.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kawata H, Kamiakito T, Omoto Y, Miyazaki C, Hozumi Y, Tanaka A. RhoC upregulation is correlated with reduced E-cadherin in human breast cancer specimens after chemotherapy and in human breast cancer MCF-7 cells. Horm cancer. 2014;5(6):414–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gou WF, Zhao Y, Lu H, Yang XF, Xiu YL, Zhao S, Liu JM, Zhu ZT, Sun HZ, Liu YP, et al. The role of RhoC in epithelial-to-mesenchymal transition of ovarian carcinoma cells. BMC Cancer. 2014;14:477.

    Article  PubMed  PubMed Central  Google Scholar 

  13. He X, Qian Y, Cai H, Yang S, Cai J, Wang Z. RhoC is essential in TGF-β1 induced epithelial-mesenchymal transition in cervical cancer cells. Oncol Lett. 2015;10(2):985–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Huang M, Prendergast GC. RhoB in cancer suppression. Histol Histopathol. 2006;21(2):213–8.

    CAS  PubMed  Google Scholar 

  15. Eckenstaler R, Hauke M, Benndorf RA. A current overview of RhoA, RhoB, and RhoC functions in vascular biology and pathology. Biochem Pharmacol. 2022;206:115321.

    Article  CAS  PubMed  Google Scholar 

  16. Dvorsky R, Ahmadian MR. Always look on the bright site of rho: structural implications for a conserved intermolecular interface. EMBO Rep. 2004;5(12):1130–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wittinghofer A, Vetter IR. Structure-function relationships of the G domain, a canonical switch motif. Annu Rev Biochem. 2011;80:943–71.

    Article  CAS  PubMed  Google Scholar 

  18. Kumawat A, Chakrabarty S, Kulkarni K. Nucleotide dependent switching in rho GTPase: conformational heterogeneity and competing molecular interactions. Sci Rep. 2017;7:45829.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zong H, Kaibuchi K, Quilliam LA. The insert region of RhoA is essential for rho kinase activation and cellular transformation. Mol Cell Biol. 2001;21(16):5287–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Dvorsky R, Blumenstein L, Vetter IR, Ahmadian MR. Structural insights into the interaction of ROCKI with the switch regions of RhoA. J Biol Chem. 2004;279(8):7098–104.

    Article  CAS  PubMed  Google Scholar 

  21. Roberts PJ, Mitin N, Keller PJ, Chenette EJ, Madigan JP, Currin RO, Cox AD, Wilson O, Kirschmeier P, Der CJ. Rho family GTPase modification and dependence on CAAX motif-signaled posttranslational modification. J Biol Chem. 2008;283(37):25150–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kinsella BT, Erdman RA, Maltese WA. Posttranslational modification of Ha-ras p21 by farnesyl versus geranylgeranyl isoprenoids is determined by the COOH-terminal amino acid. Proc Natl Acad Sci USA. 1991;88(20):8934–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Samuel F, Hynds DL. RHO GTPase signaling for axon extension: is prenylation important? Mol Neurobiol. 2010;42(2):133–42.

    Article  CAS  PubMed  Google Scholar 

  24. Backlund PS Jr. Post-translational processing of RhoA. Carboxyl methylation of the carboxyl-terminal prenylcysteine increases the half-life of Rhoa. J Biol Chem. 1997;272(52):33175–80.

    Article  CAS  PubMed  Google Scholar 

  25. Lebowitz PF, Casey PJ, Prendergast GC, Thissen JA. Farnesyltransferase inhibitors alter the prenylation and growth-stimulating function of RhoB. J Biol Chem. 1997;272(25):15591–4.

    Article  CAS  PubMed  Google Scholar 

  26. Du W, Lebowitz PF, Prendergast GC. Cell growth inhibition by farnesyltransferase inhibitors is mediated by gain of geranylgeranylated RhoB. Mol Cell Biol. 1999;19(3):1831–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Baron R, Fourcade E, Lajoie-Mazenc I, Allal C, Couderc B, Barbaras R, Favre G, Faye JC, Pradines A. RhoB prenylation is driven by the three carboxyl-terminal amino acids of the protein: evidenced in vivo by an anti-farnesyl cysteine antibody. Proc Natl Acad Sci USA. 2000;97(21):11626–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Schmidt SI, Blaabjerg M, Freude K, Meyer M. RhoA signaling in neurodegenerative diseases. Cells 2022, 11(9).

  29. Sawada N, Itoh H, Yamashita J, Doi K, Inoue M, Masatsugu K, Fukunaga Y, Sakaguchi S, Sone M, Yamahara K, et al. cGMP-dependent protein kinase phosphorylates and inactivates RhoA. Biochem Biophys Res Commun. 2001;280(3):798–805.

    Article  CAS  PubMed  Google Scholar 

  30. Guilluy C, Rolli-Derkinderen M, Loufrani L, Bourgé A, Henrion D, Sabourin L, Loirand G, Pacaud P. Ste20-related kinase SLK phosphorylates Ser188 of RhoA to induce vasodilation in response to angiotensin II type 2 receptor activation. Circul Res. 2008;102(10):1265–74.

    Article  CAS  Google Scholar 

  31. Gayard M, Guilluy C, Rousselle A, Viollet B, Henrion D, Pacaud P, Loirand G, Rolli-Derkinderen M. AMPK alpha 1-induced RhoA phosphorylation mediates vasoprotective effect of estradiol. Arterioscler Thromb Vasc Biol. 2011;31(11):2634–42.

    Article  CAS  PubMed  Google Scholar 

  32. Rolli-Derkinderen M, Sauzeau V, Boyer L, Lemichez E, Baron C, Henrion D, Loirand G, Pacaud P. Phosphorylation of serine 188 protects RhoA from ubiquitin/proteasome-mediated degradation in vascular smooth muscle cells. Circul Res. 2005;96(11):1152–60.

    Article  CAS  Google Scholar 

  33. Tong J, Li L, Ballermann B, Wang Z. Phosphorylation and activation of RhoA by ERK in response to epidermal growth factor stimulation. PLoS ONE. 2016;11(1):e0147103.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Tang J, Ip JP, Ye T, Ng YP, Yung WH, Wu Z, Fang W, Fu AK, Ip NY. Cdk5-dependent Mst3 phosphorylation and activity regulate neuronal migration through RhoA inhibition. J Neuroscience: Official J Soc Neurosci. 2014;34(22):7425–36.

    Article  CAS  Google Scholar 

  35. Liu J, Li S, Chen S, Chen S, Geng Q, Xu D. c-Met-dependent phosphorylation of RhoA plays a key role in gastric cancer tumorigenesis. J Pathol. 2019;249(1):126–36.

    Article  CAS  PubMed  Google Scholar 

  36. Kim JG, Mahmud S, Min JK, Lee YB, Kim H, Kang DC, Park HS, Seong J, Park JB. RhoA GTPase phosphorylated at tyrosine 42 by src kinase binds to β-catenin and contributes transcriptional regulation of vimentin upon Wnt3A. Redox Biol. 2021;40:101842.

    Article  CAS  PubMed  Google Scholar 

  37. Uezu A, Okada H, Murakoshi H, del Vescovo CD, Yasuda R, Diviani D, Soderling SH. Modified SH2 domain to phototrap and identify phosphotyrosine proteins from subcellular sites within cells. Proc Natl Acad Sci USA. 2012;109(43):E2929–2938.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. de la Vega M, Burrows JF, Johnston JA. Ubiquitination: added complexity in Ras and rho family GTPase function. Small GTPases. 2011;2(4):192–201.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wu Y, Liu B, Lin W, Zhao R, Han W, Xie J. AAMP promotes colorectal cancermetastasis by suppressing SMURF2-mediatedubiquitination and degradation of RhoA. Mol Therapy Oncolytics. 2021;23:515–30.

    Article  CAS  Google Scholar 

  40. Ibeawuchi SR, Agbor LN, Quelle FW, Sigmund CD. Hypertension-causing mutations in Cullin3 protein impair RhoA Protein Ubiquitination and Augment the Association with substrate adaptors. J Biol Chem. 2015;290(31):19208–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zheng J, Shi Z, Yang P, Zhao Y, Tang W, Ye S, Xuan Z, Chen C, Shao C, Wu Q et al. ERK-Smurf1-RhoA signaling is critical for TGFβ-drived EMT and tumor metastasis. Life Sci Alliance 2022, 5(10).

  42. De Pereira B, Chern YJ, He J, Chan CH. The ubiquitin ligase RNF8 regulates rho GTPases and promotes cytoskeletal changes and motility in triple-negative breast cancer cells. FEBS Lett. 2021;595(2):241–52.

    Article  Google Scholar 

  43. Mosaddeghzadeh N, Ahmadian MR. The RHO Family GTPases: mechanisms of regulation and signaling. Cells 2021, 10(7).

  44. Pawlak G, Helfman DM. Cytoskeletal changes in cell transformation and tumorigenesis. Curr Opin Genet Dev. 2001;11(1):41–7.

    Article  CAS  PubMed  Google Scholar 

  45. Li F, Higgs HN. The mouse formin mDia1 is a potent actin nucleation factor regulated by autoinhibition. Curr Biology: CB. 2003;13(15):1335–40.

    Article  CAS  Google Scholar 

  46. Higashida C, Miyoshi T, Fujita A, Oceguera-Yanez F, Monypenny J, Andou Y, Narumiya S, Watanabe N. Actin polymerization-driven molecular movement of mDia1 in living cells. Sci (New York NY). 2004;303(5666):2007–10.

    Article  CAS  Google Scholar 

  47. Narumiya S, Yasuda S. Rho GTPases in animal cell mitosis. Curr Opin Cell Biol. 2006;18(2):199–205.

    Article  CAS  PubMed  Google Scholar 

  48. Julian L, Olson MF. Rho-associated coiled-coil containing kinases (ROCK): structure, regulation, and functions. Small GTPases. 2014;5:e29846.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Clayton NS, Ridley AJ. Targeting rho GTPase signaling networks in Cancer. Front cell Dev Biology. 2020;8:222.

    Article  Google Scholar 

  50. Kimura K, Ito M, Amano M, Chihara K, Fukata Y, Nakafuku M, Yamamori B, Feng J, Nakano T, Okawa K, et al. Regulation of myosin phosphatase by rho and rho-associated kinase (Rho-kinase). Sci (New York NY). 1996;273(5272):245–8.

    Article  CAS  Google Scholar 

  51. Jaffe AB, Hall A. RHO GTPASES: Biochemistry and Biology. Annu Rev Cell Dev Biol. 2005;21(1):247–69.

    Article  CAS  PubMed  Google Scholar 

  52. Jaffe AB, Hall A. Rho GTPases in transformation and metastasis. Adv Cancer Res. 2002;84:57–80.

    Article  CAS  PubMed  Google Scholar 

  53. Ohashi K, Nagata K, Maekawa M, Ishizaki T, Narumiya S, Mizuno K. Rho-associated kinase ROCK activates LIM-kinase 1 by phosphorylation at threonine 508 within the activation loop. J Biol Chem. 2000;275(5):3577–82.

    Article  CAS  PubMed  Google Scholar 

  54. Sousa-Squiavinato ACM, Rocha MR, Barcellos-de-Souza P, de Souza WF, Morgado-Diaz JA. Cofilin-1 signaling mediates epithelial-mesenchymal transition by promoting actin cytoskeleton reorganization and cell-cell adhesion regulation in colorectal cancer cells. Biochim et Biophys acta Mol cell Res. 2019;1866(3):418–29.

    Article  CAS  Google Scholar 

  55. Maekawa M, Ishizaki T, Boku S, Watanabe N, Fujita A, Iwamatsu A, Obinata T, Ohashi K, Mizuno K, Narumiya S. Signaling from rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Sci (New York NY). 1999;285(5429):895–8.

    Article  CAS  Google Scholar 

  56. Villalonga P, Ridley AJ. Rho GTPases and cell cycle control. Growth Factors (Chur Switzerland). 2006;24(3):159–64.

    Article  CAS  PubMed  Google Scholar 

  57. Olson MF, Ashworth A, Hall A. An essential role for rho, Rac, and Cdc42 GTPases in cell cycle progression through G1. Sci (New York NY). 1995;269(5228):1270–2.

    Article  CAS  Google Scholar 

  58. Coleman ML, Marshall CJ, Olson MF. RAS and RHO GTPases in G1-phase cell-cycle regulation. Nat Rev Mol Cell Biol. 2004;5(5):355–66.

    Article  CAS  PubMed  Google Scholar 

  59. Welsh CF, Roovers K, Villanueva J, Liu Y, Schwartz MA, Assoian RK. Timing of cyclin D1 expression within G1 phase is controlled by rho. Nat Cell Biol. 2001;3(11):950–7.

    Article  CAS  PubMed  Google Scholar 

  60. Swant JD, Rendon BE, Symons M, Mitchell RA. Rho GTPase-dependent signaling is required for macrophage migration inhibitory factor-mediated expression of cyclin D1. J Biol Chem. 2005;280(24):23066–72.

    Article  CAS  PubMed  Google Scholar 

  61. Auer KL, Park JS, Seth P, Coffey RJ, Darlington G, Abo A, McMahon M, Depinho RA, Fisher PB, Dent P. Prolonged activation of the mitogen-activated protein kinase pathway promotes DNA synthesis in primary hepatocytes from p21Cip-1/WAF1-null mice, but not in hepatocytes from p16INK4a-null mice. Biochem J. 1998;336(Pt 3):551–60. (Pt 3)(.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mammoto A, Huang S, Moore K, Oh P, Ingber DE. Role of RhoA, mDia, and ROCK in cell shape-dependent control of the Skp2-p27kip1 pathway and the G1/S transition. J Biol Chem. 2004;279(25):26323–30.

    Article  CAS  PubMed  Google Scholar 

  63. Lai JM, Wu S, Huang DY, Chang ZF. Cytosolic retention of phosphorylated extracellular signal-regulated kinase and a rho-associated kinase-mediated signal impair expression of p21(Cip1/Waf1) in phorbol 12-myristate-13- acetate-induced apoptotic cells. Mol Cell Biol. 2002;22(21):7581–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Zuckerbraun BS, Shapiro RA, Billiar TR, Tzeng E. RhoA influences the nuclear localization of extracellular signal-regulated kinases to modulate p21Waf/Cip1 expression. Circulation. 2003;108(7):876–81.

    Article  CAS  PubMed  Google Scholar 

  65. Chircop M. Rho GTPases as regulators of mitosis and cytokinesis in mammalian cells. Small GTPases 2014, 5.

  66. Narumiya S, Oceguera-Yanez F, Yasuda S. A new look at rho GTPases in cell cycle: role in kinetochore-microtubule attachment. Cell Cycle (Georgetown Tex). 2004;3(7):855–7.

    Article  CAS  PubMed  Google Scholar 

  67. Oceguera-Yanez F, Narumiya S. Measurement of activity of rho GTPases during mitosis. Methods Enzymol. 2006;406:332–45.

    Article  CAS  PubMed  Google Scholar 

  68. Ferreira LT, Maiato H. Prometaphase. Semin Cell Dev Biol. 2021;117:52–61.

    Article  CAS  PubMed  Google Scholar 

  69. Glotzer M. Animal cell cytokinesis. Annu Rev Cell Dev Biol. 2001;17:351–86.

    Article  CAS  PubMed  Google Scholar 

  70. Maddox AS, Burridge K. RhoA is required for cortical retraction and rigidity during mitotic cell rounding. J Cell Biol. 2003;160(2):255–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Matthews HK, Delabre U, Rohn JL, Guck J, Kunda P, Baum B. Changes in Ect2 localization couple actomyosin-dependent cell shape changes to mitotic progression. Dev Cell. 2012;23(2):371–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Koh SP, Pham NP, Piekny A. Seeing is believing: tools to study the role of rho GTPases during cytokinesis. Small GTPases. 2022;13(1):211–24.

    Article  CAS  PubMed  Google Scholar 

  73. Kanada M, Nagasaki A, Uyeda TQ. Stabilization of anaphase midzone microtubules is regulated by rho during cytokinesis in human fibrosarcoma cells. Exp Cell Res. 2009;315(16):2705–14.

    Article  CAS  PubMed  Google Scholar 

  74. Piekny A, Werner M, Glotzer M. Cytokinesis: welcome to the rho zone. Trends Cell Biol. 2005;15(12):651–8.

    Article  CAS  PubMed  Google Scholar 

  75. Glotzer M. The molecular requirements for cytokinesis. Sci (New York NY). 2005;307(5716):1735–9.

    Article  CAS  Google Scholar 

  76. De Santis Puzzonia M, Cozzolino AM, Grassi G, Bisceglia F, Strippoli R, Guarguaglini G, Citarella F, Sacchetti B, Tripodi M, Marchetti A, et al. TGFbeta induces Binucleation/Polyploidization in Hepatocytes through a src-dependent cytokinesis failure. PLoS ONE. 2016;11(11):e0167158.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Margall-Ducos G, Celton-Morizur S, Couton D, Brégerie O, Desdouets C. Liver tetraploidization is controlled by a new process of incomplete cytokinesis. J Cell Sci. 2007;120(Pt 20):3633–9.

    Article  CAS  PubMed  Google Scholar 

  78. Goto H, Kosako H, Tanabe K, Yanagida M, Sakurai M, Amano M, Kaibuchi K, Inagaki M. Phosphorylation of vimentin by rho-associated kinase at a unique amino-terminal site that is specifically phosphorylated during cytokinesis. J Biol Chem. 1998;273(19):11728–36.

    Article  CAS  PubMed  Google Scholar 

  79. Goto H, Kosako H, Inagaki M. Regulation of intermediate filament organization during cytokinesis: possible roles of rho-associated kinase. Microsc Res Tech. 2000;49(2):173–82.

    Article  CAS  PubMed  Google Scholar 

  80. Heib T, Hermanns HM, Manukjan G, Englert M, Kusch C, Becker IC, Gerber A, Wackerbarth LM, Burkard P, Dandekar T, et al. RhoA/Cdc42 signaling drives cytoplasmic maturation but not endomitosis in megakaryocytes. Cell Rep. 2021;35(6):109102.

    Article  CAS  PubMed  Google Scholar 

  81. Mazzi S, Lordier L, Debili N, Raslova H, Vainchenker W. Megakaryocyte and polyploidization. Exp Hematol. 2018;57:1–13.

    Article  CAS  PubMed  Google Scholar 

  82. Lordier L, Jalil A, Aurade F, Larbret F, Larghero J, Debili N, Vainchenker W, Chang Y. Megakaryocyte endomitosis is a failure of late cytokinesis related to defects in the contractile ring and Rho/Rock signaling. Blood. 2008;112(8):3164–74.

    Article  CAS  PubMed  Google Scholar 

  83. Green RA, Paluch E, Oegema K. Cytokinesis in animal cells. Annu Rev Cell Dev Biol. 2012;28:29–58.

    Article  CAS  PubMed  Google Scholar 

  84. Lu C, Mahajan A, Hong SH, Galli S, Zhu S, Tilan JU, Abualsaud N, Adnani M, Chung S, Elmansy N, et al. Hypoxia-activated neuropeptide Y/Y5 receptor/RhoA pathway triggers chromosomal instability and bone metastasis in ewing sarcoma. Nat Commun. 2022;13(1):2323.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Taneja N, Baillargeon SM, Burnette DT. Myosin light chain kinase-driven myosin II turnover regulates actin cortex contractility during mitosis. Mol Biol Cell. 2021;32(20):br3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Cheffings TH, Burroughs NJ, Balasubramanian MK. Actin turnover ensures uniform tension distribution during cytokinetic actomyosin ring contraction. Mol Biol Cell. 2019;30(8):933–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Malla M, Pollard TD, Chen Q. Counting actin in contractile rings reveals novel contributions of cofilin and type II myosins to fission yeast cytokinesis. Mol Biol Cell. 2022;33(6):ar51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Nagaoka R, Abe H, Kusano K, Obinata T. Concentration of cofilin, a small actin-binding protein, at the cleavage furrow during cytokinesis. Cell Motil Cytoskeleton. 1995;30(1):1–7.

    Article  CAS  PubMed  Google Scholar 

  89. McDargh Z, Zhu T, Zhu H, O’Shaughnessy B. Actin turnover protects the cytokinetic contractile ring from structural instability. J Cell Sci 2023, 136(5).

  90. Woolner S, Bement WM. Unconventional myosins acting unconventionally. Trends Cell Biol. 2009;19(6):245–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Kalendová A, Kalasová I, Yamazaki S, Uličná L, Harata M, Hozák P. Nuclear actin filaments recruit cofilin and actin-related protein 3, and their formation is connected with a mitotic block. Histochem Cell Biol. 2014;142(2):139–52.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Baarlink C, Plessner M, Sherrard A, Morita K, Misu S, Virant D, Kleinschnitz EM, Harniman R, Alibhai D, Baumeister S, et al. A transient pool of nuclear F-actin at mitotic exit controls chromatin organization. Nat Cell Biol. 2017;19(12):1389–99.

    Article  CAS  PubMed  Google Scholar 

  93. Epp JA, Chant J. An IQGAP-related protein controls actin-ring formation and cytokinesis in yeast. Curr Biology: CB. 1997;7(12):921–9.

    Article  CAS  PubMed  Google Scholar 

  94. Adachi H, Takahashi Y, Hasebe T, Shirouzu M, Yokoyama S, Sutoh K. Dictyostelium IQGAP-related protein specifically involved in the completion of cytokinesis. J Cell Biol. 1997;137(4):891–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Adachi M, Kawasaki A, Nojima H, Nishida E, Tsukita S. Involvement of IQGAP family proteins in the regulation of mammalian cell cytokinesis. Genes Cells: Devoted Mol Cell Mech. 2014;19(11):803–20.

    Article  CAS  Google Scholar 

  96. David MD, Petit D, Bertoglio J. The RhoGAP ARHGAP19 controls cytokinesis and chromosome segregation in T lymphocytes. J Cell Sci. 2014;127(Pt 2):400–10.

    CAS  PubMed  Google Scholar 

  97. Marceaux C, Petit D, Bertoglio J, David MD. Phosphorylation of ARHGAP19 by CDK1 and ROCK regulates its subcellular localization and function during mitosis. J Cell Sci 2018, 131(5).

  98. Quadri R, Rotondo G, Sertic S, Pozzi S, dell’Oca MC, Guerrini L, Muzi-Falconi M. A Haspin-ARHGAP11A axis regulates epithelial morphogenesis through Rho-ROCK dependent modulation of LIMK1-Cofilin. iScience 2023, 26(10):108011.

  99. Davies T, Canman JC. Stuck in the middle: Rac, adhesion, and cytokinesis. J Cell Biol. 2012;198(5):769–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Baumann K. Cell cycle: centralspindlin–the missing link. Nat Rev Mol Cell Biol. 2013;14(2):68.

    Article  CAS  PubMed  Google Scholar 

  101. Lorès P, Vernet N, Kurosaki T, Van de Putte T, Huylebroeck D, Hikida M, Gacon G, Touré A. Deletion of MgcRacGAP in the male germ cells impairs spermatogenesis and causes male sterility in the mouse. Dev Biol. 2014;386(2):419–27.

    Article  PubMed  Google Scholar 

  102. Tatsumoto T, Xie X, Blumenthal R, Okamoto I, Miki T. Human ECT2 is an exchange factor for rho GTPases, phosphorylated in G2/M phases, and involved in cytokinesis. J Cell Biol. 1999;147(5):921–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Kamijo K, Ohara N, Abe M, Uchimura T, Hosoya H, Lee JS, Miki T. Dissecting the role of rho-mediated signaling in contractile ring formation. Mol Biol Cell. 2006;17(1):43–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Benais-Pont G, Punn A, Flores-Maldonado C, Eckert J, Raposo G, Fleming TP, Cereijido M, Balda MS, Matter K. Identification of a tight junction-associated guanine nucleotide exchange factor that activates rho and regulates paracellular permeability. J Cell Biol. 2003;160(5):729–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Bakal CJ, Finan D, LaRose J, Wells CD, Gish G, Kulkarni S, DeSepulveda P, Wilde A, Rottapel R. The rho GTP exchange factor Lfc promotes spindle assembly in early mitosis. Proc Natl Acad Sci USA. 2005;102(27):9529–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Kwan KM, Kirschner MW. A microtubule-binding Rho-GEF controls cell morphology during convergent extension of Xenopus laevis. Development. 2005;132(20):4599–610.

    Article  CAS  PubMed  Google Scholar 

  107. Zanin E, Desai A, Poser I, Toyoda Y, Andree C, Moebius C, Bickle M, Conradt B, Piekny A, Oegema K. A conserved RhoGAP limits M phase contractility and coordinates with microtubule asters to confine RhoA during cytokinesis. Dev Cell. 2013;26(5):496–510.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Sharma S, Rikhy R. Spatiotemporal recruitment of RhoGTPase protein GRAF inhibits actomyosin ring constriction in Drosophila cellularization. eLife 2021, 10.

  109. Mikawa M, Su L, Parsons SJ. Opposing roles of p190RhoGAP and Ect2 RhoGEF in regulating cytokinesis. Cell Cycle (Georgetown Tex). 2008;7(13):2003–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Manukyan A, Ludwig K, Sanchez-Manchinelly S, Parsons SJ, Stukenberg PT. A complex of p190RhoGAP-A and anillin modulates RhoA-GTP and the cytokinetic furrow in human cells. J Cell Sci. 2015;128(1):50–60.

    CAS  PubMed  Google Scholar 

  111. Agircan FG, Schiebel E, Mardin BR. Separate to operate: control of centrosome positioning and separation. Philosophical Trans Royal Soc Lond Ser B Biol Sci 2014, 369(1650).

  112. Blanco-Ameijeiras J, Lozano-Fernández P, Martí E. Centrosome maturation - in tune with the cell cycle. J Cell Sci 2022, 135(2).

  113. Tsou MF, Stearns T. Mechanism limiting centrosome duplication to once per cell cycle. Nature. 2006;442(7105):947–51.

    Article  CAS  PubMed  Google Scholar 

  114. Firdous F, Raza HG, Chotana GA, Choudhary MI, Faisal A, Saleem RSZ. Centrosome Clustering & Chemotherapy. Mini Rev Med Chem. 2023;23(4):429–51.

    Article  CAS  PubMed  Google Scholar 

  115. Sir JH, Pütz M, Daly O, Morrison CG, Dunning M, Kilmartin JV, Gergely F. Loss of centrioles causes chromosomal instability in vertebrate somatic cells. J Cell Biol. 2013;203(5):747–56.

    Article  CAS  PubMed  Google Scholar 

  116. Holland AJ, Cleveland DW. Losing balance: the origin and impact of aneuploidy in cancer. EMBO Rep. 2012;13(6):501–14.

    Article  CAS  PubMed  Google Scholar 

  117. Arquint C, Gabryjonczyk AM, Nigg EA. Centrosomes as signalling centres. Philosophical Trans Royal Soc Lond Ser B Biol Sci 2014, 369(1650).

  118. Ma Z, Kanai M, Kawamura K, Kaibuchi K, Ye K, Fukasawa K. Interaction between ROCK II and nucleophosmin/B23 in the regulation of centrosome duplication. Mol Cell Biol. 2006;26(23):9016–34.

    Article  CAS  PubMed  Google Scholar 

  119. Wang HF, Takenaka K, Nakanishi A, Miki Y. BRCA2 and nucleophosmin coregulate centrosome amplification and form a complex with the rho effector kinase ROCK2. Cancer Res. 2011;71(1):68–77.

    Article  CAS  PubMed  Google Scholar 

  120. Chevrier V, Piel M, Collomb N, Saoudi Y, Frank R, Paintrand M, Narumiya S, Bornens M, Job D. The rho-associated protein kinase p160ROCK is required for centrosome positioning. J Cell Biol. 2002;157(5):807–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Kloc M, Uosef A, Wosik J, Kubiak JZ, Ghobrial RM. RhoA pathway and actin regulation of the Golgi/Centriole complex. Result Probl Cell Differ. 2019;67:81–93.

    Article  CAS  Google Scholar 

  122. Oku Y, Tareyanagi C, Takaya S, Osaka S, Ujiie H, Yoshida K, Nishiya N, Uehara Y. Multimodal effects of small molecule ROCK and LIMK inhibitors on mitosis, and their implication as anti-leukemia agents. PLoS ONE. 2014;9(3):e92402.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Rosenblatt J, Cramer LP, Baum B, McGee KM. Myosin II-dependent cortical movement is required for centrosome separation and positioning during mitotic spindle assembly. Cell. 2004;117(3):361–72.

    Article  CAS  PubMed  Google Scholar 

  124. Hwang M, Peddibhotla S, McHenry P, Chang P, Yochum Z, Park KU, Sears JC, Vargo-Gogola T. P190B RhoGAP regulates chromosome segregation in Cancer cells. Cancers. 2012;4(2):475–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. White EA, Glotzer M. Centralspindlin: at the heart of cytokinesis. Cytoskeleton (Hoboken NJ). 2012;69(11):882–92.

    Article  CAS  Google Scholar 

  126. Aoki T, Ueda S, Kataoka T, Satoh T. Regulation of mitotic spindle formation by the RhoA guanine nucleotide exchange factor ARHGEF10. BMC Cell Biol. 2009;10:56.

    Article  PubMed  Google Scholar 

  127. Duan X, Sun SC. Actin cytoskeleton dynamics in mammalian oocyte meiosis. Biol Reprod. 2019;100(1):15–24.

    Article  PubMed  Google Scholar 

  128. Sun QY, Schatten H. Regulation of dynamic events by microfilaments during oocyte maturation and fertilization. Reprod (Cambridge England). 2006;131(2):193–205.

    Article  CAS  Google Scholar 

  129. Bourdais A, Dehapiot B, Halet G. Cofilin regulates actin network homeostasis and microvilli length in mouse oocytes. J Cell Sci 2021, 134(24).

  130. Jin ZL, Jo YJ, Namgoong S, Kim NH. CAP1-mediated actin cycling via ADF/cofilin proteins is essential for asymmetric division in mouse oocytes. J Cell Sci 2018, 131(23).

  131. Murali A, Rajalingam K. Small rho GTPases in the control of cell shape and mobility. Cell Mol Life Sci. 2014;71(9):1703–21.

    Article  CAS  PubMed  Google Scholar 

  132. Braga VM, Yap AS. The challenges of abundance: epithelial junctions and small GTPase signalling. Curr Opin Cell Biol. 2005;17(5):466–74.

    Article  CAS  PubMed  Google Scholar 

  133. Pertz O, Hodgson L, Klemke RL, Hahn KM. Spatiotemporal dynamics of RhoA activity in migrating cells. Nature. 2006;440(7087):1069–72.

    Article  CAS  PubMed  Google Scholar 

  134. Alblas J, Ulfman L, Hordijk P, Koenderman L. Activation of Rhoa and ROCK are essential for detachment of migrating leukocytes. Mol Biol Cell. 2001;12(7):2137–45.

    Article  CAS  PubMed  Google Scholar 

  135. Ridley AJ. Rho GTPases and cell migration. J Cell Sci. 2001;114(15):2713–22.

    Article  CAS  PubMed  Google Scholar 

  136. Aizawa H, Sutoh K, Yahara I. Overexpression of cofilin stimulates bundling of actin filaments, membrane ruffling, and cell movement in Dictyostelium. J Cell Biol. 1996;132(3):335–44.

    Article  CAS  PubMed  Google Scholar 

  137. Kurokawa K, Matsuda M. Localized RhoA activation as a requirement for the induction of membrane ruffling. Mol Biol Cell. 2005;16(9):4294–303.

    Article  CAS  PubMed  Google Scholar 

  138. Dasgupta SK, Le A, Vijayan KV, Thiagarajan P. Dasatinib inhibits actin fiber reorganization and promotes endothelial cell permeability through RhoA-ROCK pathway. Cancer Med. 2017;6(4):809–18.

    Article  CAS  PubMed  Google Scholar 

  139. Mu G, Ding Q, Li H, Zhang L, Zhang L, He K, Wu L, Deng Y, Yang D, Wu L, et al. Gastrin stimulates pancreatic cancer cell directional migration by activating the Gα12/13-RhoA-ROCK signaling pathway. Exp Mol Med. 2018;50(5):1–14.

    Article  CAS  PubMed  Google Scholar 

  140. Rottner K, Hall A, Small JV. Interplay between Rac and rho in the control of substrate contact dynamics. Curr Biology: CB. 1999;9(12):640–8.

    Article  CAS  PubMed  Google Scholar 

  141. Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28(1–2):15–33.

    Article  PubMed  Google Scholar 

  142. Shankar J, Nabi IR. Actin cytoskeleton regulation of epithelial mesenchymal transition in metastatic cancer cells. PLoS ONE. 2015;10(3):e0119954.

    Article  PubMed  Google Scholar 

  143. Charras GT, Yarrow JC, Horton MA, Mahadevan L, Mitchison TJ. Non-equilibration of hydrostatic pressure in blebbing cells. Nature. 2005;435(7040):365–9.

    Article  CAS  PubMed  Google Scholar 

  144. Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3(5):362–74.

    Article  CAS  PubMed  Google Scholar 

  145. Friedl P, Bröcker EB. The biology of cell locomotion within three-dimensional extracellular matrix. Cell Mol Life Sci. 2000;57(1):41–64.

    Article  CAS  PubMed  Google Scholar 

  146. Linder S, Wiesner C, Himmel M. Degrading devices: invadosomes in proteolytic cell invasion. Annu Rev Cell Dev Biol. 2011;27:185–211.

    Article  CAS  PubMed  Google Scholar 

  147. Jeong KJ, Park SY, Cho KH, Sohn JS, Lee J, Kim YK, Kang J, Park CG, Han JW, Lee HY. The Rho/ROCK pathway for lysophosphatidic acid-induced proteolytic enzyme expression and ovarian cancer cell invasion. Oncogene. 2012;31(39):4279–89.

    Article  CAS  PubMed  Google Scholar 

  148. Zhang JG, Li XY, Wang YZ, Zhang QD, Gu SY, Wu X, Zhu GH, Li Q, Liu GL. ROCK is involved in vasculogenic mimicry formation in hepatocellular carcinoma cell line. PLoS ONE. 2014;9(9):e107661.

    Article  PubMed  Google Scholar 

  149. Pagano C, Navarra G, Pastorino O, Avilia G, Coppola L, Della Monica R, Chiariotti L, Florio T, Corsaro A, Torelli G et al. N6-Isopentenyladenosine Hinders the Vasculogenic Mimicry in Human Glioblastoma Cells through Src-120 Catenin Pathway Modulation and RhoA Activity Inhibition. International journal of molecular sciences 2021, 22(19).

  150. Zhang JG, Zhou HM, Zhang X, Mu W, Hu JN, Liu GL, Li Q. Hypoxic induction of vasculogenic mimicry in hepatocellular carcinoma: role of HIF-1 α, RhoA/ROCK and Rac1/PAK signaling. BMC Cancer. 2020;20(1):32.

    Article  PubMed  Google Scholar 

  151. Xia Y, Cai XY, Fan JQ, Zhang LL, Ren JH, Li ZY, Zhang RG, Zhu F, Wu G. The role of sema4D in vasculogenic mimicry formation in non-small cell lung cancer and the underlying mechanisms. Int J Cancer. 2019;144(9):2227–38.

    Article  CAS  PubMed  Google Scholar 

  152. Zhang X, Zhang J, Zhou H, Liu G, Li Q. Rho kinase mediates transforming growth factor-β1-induced vasculogenic mimicry formation: involvement of the epithelial-mesenchymal transition and cancer stemness activity. Acta Biochim Biophys Sin. 2020;52(4):411–20.

    Article  CAS  PubMed  Google Scholar 

  153. Zhang Z, Nong L, Chen M, Gu X, Zhao W, Liu M, Cheng W. Baicalein suppresses vasculogenic mimicry through inhibiting RhoA/ROCK expression in lung cancer A549 cell line. Acta Biochim Biophys Sin. 2020;52(9):1007–15.

    Article  CAS  Google Scholar 

  154. Zhang JG, Zhang DD, Wu X, Wang YZ, Gu SY, Zhu GH, Li XY, Li Q, Liu GL. Incarvine C suppresses proliferation and vasculogenic mimicry of hepatocellular carcinoma cells via targeting ROCK inhibition. BMC Cancer. 2015;15:814.

    Article  PubMed  Google Scholar 

  155. Ebrahimi N, Afshinpour M, Fakhr SS, Kalkhoran PG, Shadman-Manesh V, Adelian S, Beiranvand S, Rezaei-Tazangi F, Khorram R, Hamblin MR et al. Cancer stem cells in colorectal cancer: Signaling pathways involved in stemness and therapy resistance. Critical reviews in oncology/hematology 2023, 182:103920.

  156. Uribe-Etxebarria V, Pineda JR, García-Gallastegi P, Agliano A, Unda F, Ibarretxe G. Notch and wnt signaling modulation to Enhance DPSC stemness and therapeutic potential. Int J Mol Sci 2023, 24(8).

  157. Ebrahimi N, Kharazmi K, Ghanaatian M, Miraghel SA, Amiri Y, Seyedebrahimi SS, Mobarak H, Yazdani E, Parkhideh S, Hamblin MR, et al. Role of the wnt and GTPase pathways in breast cancer tumorigenesis and treatment. Cytokine Growth Factor Rev. 2022;67:11–24.

    Article  CAS  PubMed  Google Scholar 

  158. Kobune M, Chiba H, Kato J, Kato K, Nakamura K, Kawano Y, Takada K, Takimoto R, Takayama T, Hamada H, et al. Wnt3/RhoA/ROCK signaling pathway is involved in adhesion-mediated drug resistance of multiple myeloma in an autocrine mechanism. Mol Cancer Ther. 2007;6(6):1774–84.

    Article  CAS  PubMed  Google Scholar 

  159. Kim KB, Kim DW, Kim Y, Tang J, Kirk N, Gan Y, Kim B, Fang B, Park JL, Zheng Y, et al. WNT5A-RHOA signaling is a driver of Tumorigenesis and represents a therapeutically actionable vulnerability in small cell Lung Cancer. Cancer Res. 2022;82(22):4219–33.

    Article  CAS  PubMed  Google Scholar 

  160. Zanconato F, Cordenonsi M, Piccolo S. YAP/TAZ at the roots of Cancer. Cancer Cell. 2016;29(6):783–803.

    Article  CAS  PubMed  Google Scholar 

  161. Yan Z, Guo D, Tao R, Yu X, Zhang J, He Y, Zhang J, Li J, Zhang S, Guo W. Fluid shear stress induces cell migration via RhoA-YAP1-autophagy pathway in liver cancer stem cells. Cell Adhes Migr. 2022;16(1):94–106.

    Article  CAS  Google Scholar 

  162. Fujimoto D, Ueda Y, Hirono Y, Goi T, Yamaguchi A. PAR1 participates in the ability of multidrug resistance and tumorigenesis by controlling Hippo-YAP pathway. Oncotarget. 2015;6(33):34788–99.

    Article  PubMed  Google Scholar 

  163. Zheng L, Xiang C, Li X, Guo Q, Gao L, Ni H, Xia Y, Xi T. STARD13-correlated ceRNA network-directed inhibition on YAP/TAZ activity suppresses stemness of breast cancer via co-regulating Hippo and Rho-GTPase/F-actin signaling. J Hematol Oncol. 2018;11(1):72.

    Article  PubMed  Google Scholar 

  164. Alkasalias T, Alexeyenko A, Hennig K, Danielsson F, Lebbink RJ, Fielden M, Turunen SP, Lehti K, Kashuba V, Madapura HS, et al. RhoA knockout fibroblasts lose tumor-inhibitory capacity in vitro and promote tumor growth in vivo. Proc Natl Acad Sci USA. 2017;114(8):E1413–21.

    Article  CAS  PubMed  Google Scholar 

  165. Saygin C, Matei D, Majeti R, Reizes O, Lathia JD. Targeting Cancer Stemness in the clinic: from hype to Hope. Cell Stem Cell. 2019;24(1):25–40.

    Article  CAS  PubMed  Google Scholar 

  166. Zhang S, Mercado-Uribe I, Liu J. Tumor stroma and differentiated cancer cells can be originated directly from polyploid giant cancer cells induced by paclitaxel. Int J Cancer. 2014;134(3):508–18.

    Article  CAS  PubMed  Google Scholar 

  167. Zhang S, Mercado-Uribe I, Xing Z, Sun B, Kuang J, Liu J. Generation of cancer stem-like cells through the formation of polyploid giant cancer cells. Oncogene. 2014;33(1):116–28.

    Article  CAS  PubMed  Google Scholar 

  168. Zhou X, Zhou M, Zheng M, Tian S, Yang X, Ning Y, Li Y, Zhang S. Polyploid giant cancer cells and cancer progression. Front cell Dev Biology. 2022;10:1017588.

    Article  Google Scholar 

  169. Erenpreisa J, Kalejs M, Ianzini F, Kosmacek EA, Mackey MA, Emzinsh D, Cragg MS, Ivanov A, Illidge TM. Segregation of genomes in polyploid tumour cells following mitotic catastrophe. Cell Biol Int. 2005;29(12):1005–11.

    Article  CAS  PubMed  Google Scholar 

  170. Vitale I, Galluzzi L, Senovilla L, Criollo A, Jemaà M, Castedo M, Kroemer G. Illicit survival of cancer cells during polyploidization and depolyploidization. Cell Death Differ. 2011;18(9):1403–13.

    Article  CAS  PubMed  Google Scholar 

  171. Was H, Borkowska A, Olszewska A, Klemba A, Marciniak M, Synowiec A, Kieda C. Polyploidy formation in cancer cells: how a trojan horse is born. Sem Cancer Biol 2021.

  172. Xuan B, Ghosh D, Cheney EM, Clifton EM, Dawson MR. Dysregulation in Actin Cytoskeletal Organization Drives Increased Stiffness and migratory persistence in Polyploidal Giant Cancer cells. Sci Rep. 2018;8(1):11935.

    Article  PubMed  Google Scholar 

  173. Batlle E, Clevers H. Cancer stem cells revisited. Nat Med. 2017;23(10):1124–34.

    Article  CAS  PubMed  Google Scholar 

  174. Phan TG, Croucher PI. The dormant cancer cell life cycle. Nat Rev Cancer. 2020;20(7):398–411.

    Article  CAS  PubMed  Google Scholar 

  175. Yoon C, Cho SJ, Aksoy BA, Park DJ, Schultz N, Ryeom SW, Yoon SS. Chemotherapy resistance in diffuse-type gastric adenocarcinoma is mediated by RhoA activation in Cancer Stem-Like cells. Clin cancer Research: Official J Am Association Cancer Res. 2016;22(4):971–83.

    Article  CAS  Google Scholar 

  176. Szakács G, Annereau JP, Lababidi S, Shankavaram U, Arciello A, Bussey KJ, Reinhold W, Guo Y, Kruh GD, Reimers M, et al. Predicting drug sensitivity and resistance: profiling ABC transporter genes in cancer cells. Cancer Cell. 2004;6(2):129–37.

    Article  PubMed  Google Scholar 

  177. Gillet JP, Efferth T, Steinbach D, Hamels J, de Longueville F, Bertholet V, Remacle J. Microarray-based detection of multidrug resistance in human tumor cells by expression profiling of ATP-binding cassette transporter genes. Cancer Res. 2004;64(24):8987–93.

    Article  CAS  PubMed  Google Scholar 

  178. Huang Y, Sadée W. Membrane transporters and channels in chemoresistance and -sensitivity of tumor cells. Cancer Lett. 2006;239(2):168–82.

    Article  CAS  PubMed  Google Scholar 

  179. Chen ZX, Pervaiz S. Bcl-2 induces pro-oxidant state by engaging mitochondrial respiration in tumor cells. Cell Death Differ. 2007;14(9):1617–27.

    Article  CAS  PubMed  Google Scholar 

  180. Brenner C, Cadiou H, Vieira HL, Zamzami N, Marzo I, Xie Z, Leber B, Andrews D, Duclohier H, Reed JC, et al. Bcl-2 and Bax regulate the channel activity of the mitochondrial adenine nucleotide translocator. Oncogene. 2000;19(3):329–36.

    Article  CAS  PubMed  Google Scholar 

  181. Sau A, Pellizzari Tregno F, Valentino F, Federici G, Caccuri AM. Glutathione transferases and development of new principles to overcome drug resistance. Arch Biochem Biophys. 2010;500(2):116–22.

    Article  CAS  PubMed  Google Scholar 

  182. Zhang S, Chatterjee T, Godoy C, Wu L, Liu QJ, Carmon KS. GPR56 drives colorectal tumor growth and promotes Drug Resistance through Upregulation of MDR1 expression via a RhoA-Mediated mechanism. Mol cancer Research: MCR. 2019;17(11):2196–207.

    Article  CAS  PubMed  Google Scholar 

  183. Doublier S, Riganti C, Voena C, Costamagna C, Aldieri E, Pescarmona G, Ghigo D, Bosia A. RhoA silencing reverts the resistance to doxorubicin in human colon cancer cells. Mol cancer Research: MCR. 2008;6(10):1607–20.

    Article  CAS  PubMed  Google Scholar 

  184. Ruihua H, Mengyi Z, Chong Z, Meng Q, Xin M, Qiulin T, Feng B, Ming L. RhoA regulates resistance to irinotecan by regulating membrane transporter and apoptosis signaling in colorectal cancer. Oncotarget. 2016;7(52):87136–46.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Shen DW, Pouliot LM, Gillet JP, Ma W, Johnson AC, Hall MD, Gottesman MM. The transcription factor GCF2 is an upstream repressor of the small GTPAse RhoA, regulating membrane protein trafficking, sensitivity to doxorubicin, and resistance to cisplatin. Mol Pharm. 2012;9(6):1822–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Shen DW, Su A, Liang XJ, Pai-Panandiker A, Gottesman MM. Reduced expression of small GTPases and hypermethylation of the folate binding protein gene in cisplatin-resistant cells. Br J Cancer. 2004;91(2):270–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Zhan F, He T, Chen Z, Zuo Q, Wang Y, Li Q, Zhong S, Ou Y. RhoA enhances osteosarcoma resistance to MPPa-PDT via the Hippo/YAP signaling pathway. Cell Bioscience. 2021;11(1):179.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Yin CP, Guan SH, Zhang B, Wang XX, Yue SW. Upregulation of HIF-1α protects neuroblastoma cells from hypoxia-induced apoptosis in a RhoA-dependent manner. Mol Med Rep. 2015;12(5):7123–31.

    Article  CAS  PubMed  Google Scholar 

  189. Adua SJ, Arnal-Estapé A, Zhao M, Qi B, Liu ZZ, Kravitz C, Hulme H, Strittmatter N, López-Giráldez F, Chande S, et al. Brain metastatic outgrowth and osimertinib resistance are potentiated by RhoA in EGFR-mutant lung cancer. Nat Commun. 2022;13(1):7690.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Chen X, Liu J, Cheng L, Li C, Zhang Z, Bai Y, Wang R, Han T, Huang C, Kong Y, et al. Inhibition of noncanonical wnt pathway overcomes enzalutamide resistance in castration-resistant prostate cancer. Prostate. 2020;80(3):256–66.

    Article  PubMed  Google Scholar 

  191. Zhou Y, Zhou Y, Wang K, Li T, Zhang M, Yang Y, Wang R, Hu R. ROCK2 Confers Acquired Gemcitabine Resistance in Pancreatic Cancer Cells by Upregulating Transcription Factor ZEB1. Cancers 2019, 11(12).

  192. Zhang X, Li T, Yang M, Du Q, Wang R, Fu B, Tan Y, Cao M, Chen Y, Wang Q, et al. Acquired temozolomide resistance in MGMT(low) gliomas is associated with regulation of homologous recombination repair by ROCK2. Cell Death Dis. 2022;13(2):138.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Gao W, Zhang S, Guorong L, Liu Q, Zhu A, Gui F, Zou Y, Wu Y, Luo Y, Hong Z. Nc886 promotes renal cancer cell drug-resistance by enhancing EMT through Rock2 phosphorylation-mediated β-catenin nuclear translocation. Cell Cycle (Georgetown Tex). 2022;21(4):340–51.

    Article  CAS  PubMed  Google Scholar 

  194. Liang L, Gu W, Li M, Gao R, Zhang X, Guo C, Mi S. The long noncoding RNA HOTAIRM1 controlled by AML1 enhances glucocorticoid resistance by activating RHOA/ROCK1 pathway through suppressing ARHGAP18. Cell Death Dis. 2021;12(7):702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Pranatharthi A, Ross C, Srivastava S. Cancer Stem Cells and Radioresistance: Rho/ROCK Pathway Plea Attention. Stem cells international 2016, 2016:5785786.

  196. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–60.

    Article  CAS  PubMed  Google Scholar 

  197. Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, Qian D, Lam JS, Ailles LE, Wong M, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458(7239):780–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Osaki JH, Espinha G, Magalhaes YT, Forti FL. Modulation of RhoA GTPase Activity Sensitizes Human Cervix Carcinoma Cells to γ-Radiation by Attenuating DNA Repair Pathways. Oxidative medicine and cellular longevity 2016, 2016:6012642.

  199. Dubash AD, Guilluy C, Srougi MC, Boulter E, Burridge K, García-Mata R. The small GTPase RhoA localizes to the nucleus and is activated by Net1 and DNA damage signals. PLoS ONE. 2011;6(2):e17380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. McLaughlin N, Annabi B, Bouzeghrane M, Temme A, Bahary JP, Moumdjian R, Béliveau R. The Survivin-mediated radioresistant phenotype of glioblastomas is regulated by RhoA and inhibited by the green tea polyphenol (-)-epigallocatechin-3-gallate. Brain Res. 2006;1071(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  201. Xia N, Yang N, Shan Q, Wang Z, Liu X, Chen Y, Lu J, Huang W, Wang Z. HNRNPC regulates RhoA to induce DNA damage repair and cancer-associated fibroblast activation causing radiation resistance in pancreatic cancer. J Cell Mol Med. 2022;26(8):2322–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Magalhaes YT, Forti FL. ROCK inhibition reduces the sensitivity of mutant p53 glioblastoma to genotoxic stress through a Rac1-driven ROS production. Int J Biochem Cell Biol. 2023;164:106474.

    Article  CAS  PubMed  Google Scholar 

  203. Zheng CW, Zeng RJ, Xu LY, Li EM. Rho GTPases: promising candidates for overcoming chemotherapeutic resistance. Cancer Lett. 2020;475:65–78.

    Article  CAS  PubMed  Google Scholar 

  204. Magalhaes YT, Cardella GD, Forti FL. Exoenzyme C3 transferase lowers actin cytoskeleton dynamics, genomic stability and survival of malignant melanoma cells under UV-light stress. J Photochem Photobiology B Biology. 2020;209:111947.

    Article  CAS  Google Scholar 

  205. Yao Y, Sun S, Cao M, Mao M, He J, Gai Q, Qin Y, Yao X, Lu H, Chen F, et al. Grincamycin B functions as a potent inhibitor for Glioblastoma Stem Cell via Targeting RHOA and PI3K/AKT. ACS Chem Neurosci. 2020;11(15):2256–65.

    Article  CAS  PubMed  Google Scholar 

  206. Tsubaki M, Genno S, Takeda T, Matsuda T, Kimura N, Yamashita Y, Morii Y, Shimomura K, Nishida S. Rhosin suppressed tumor cell metastasis through inhibition of Rho/YAP pathway and expression of RHAMM and CXCR4 in melanoma and breast Cancer cells. Biomedicines 2021, 9(1).

  207. Evelyn CR, Bell JL, Ryu JG, Wade SM, Kocab A, Harzdorf NL, Showalter HD, Neubig RR, Larsen SD. Design, synthesis and prostate cancer cell-based studies of analogs of the Rho/MKL1 transcriptional pathway inhibitor, CCG-1423. Bioorg Med Chem Lett. 2010;20(2):665–72.

    Article  CAS  PubMed  Google Scholar 

  208. Johnson LA, Rodansky ES, Haak AJ, Larsen SD, Neubig RR, Higgins PD. Novel Rho/MRTF/SRF inhibitors block matrix-stiffness and TGF-β-induced fibrogenesis in human colonic myofibroblasts. Inflamm Bowel Dis. 2014;20(1):154–65.

    Article  PubMed  Google Scholar 

  209. Kim JH, Park S, Lim SM, Eom HJ, Balch C, Lee J, Kim GJ, Jeong JH, Nam S, Kim YH. Rational design of small molecule RHOA inhibitors for gastric cancer. Pharmacogenomics J. 2020;20(4):601–12.

    Article  CAS  PubMed  Google Scholar 

  210. Yang Y, Wang S, Cai J, Liang J, Zhang Y, Xie Y, Luo F, Tang J, Gao Y, Shen S, et al. Targeting ARHGEF12 promotes neuroblastoma differentiation, MYCN degradation, and reduces tumorigenicity. Cell Oncol (Dordrecht). 2023;46(1):133–43.

    Article  CAS  Google Scholar 

  211. Takahashi N, Nobusue H, Shimizu T, Sugihara E, Yamaguchi-Iwai S, Onishi N, Kunitomi H, Kuroda T, Saya H. ROCK inhibition induces terminal adipocyte differentiation and suppresses tumorigenesis in Chemoresistant Osteosarcoma Cells. Cancer Res. 2019;79(12):3088–99.

    Article  CAS  PubMed  Google Scholar 

  212. Srinivasan S, Ashok V, Mohanty S, Das A, Das S, Kumar S, Sen S, Purwar R. Blockade of rho-associated protein kinase (ROCK) inhibits the contractility and invasion potential of cancer stem like cells. Oncotarget. 2017;8(13):21418–28.

    Article  PubMed  PubMed Central  Google Scholar 

  213. Tsai CC, Liu HF, Hsu KC, Yang JM, Chen C, Liu KK, Hsu TS, Chao JI. 7-Chloro-6-piperidin-1-yl-quinoline-5,8-dione (PT-262), a novel ROCK inhibitor blocks cytoskeleton function and cell migration. Biochem Pharmacol. 2011;81(7):856–65.

    Article  CAS  PubMed  Google Scholar 

  214. Patel RA, Forinash KD, Pireddu R, Sun Y, Sun N, Martin MP, Schönbrunn E, Lawrence NJ, Sebti SM. RKI-1447 is a potent inhibitor of the rho-associated ROCK kinases with anti-invasive and antitumor activities in breast cancer. Cancer Res. 2012;72(19):5025–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Rath N, Munro J, Cutiongco MF, Jagiełło A, Gadegaard N, McGarry L, Unbekandt M, Michalopoulou E, Kamphorst JJ, Sumpton D, et al. Rho kinase inhibition by AT13148 blocks pancreatic ductal Adenocarcinoma Invasion and Tumor Growth. Cancer Res. 2018;78(12):3321–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Kim S, Kim SA, Nam GH, Hong Y, Kim GB, Choi Y, Lee S, Cho Y, Kwon M, Jeong C et al. In situ immunogenic clearance induced by a combination of photodynamic therapy and rho-kinase inhibition sensitizes immune checkpoint blockade response to elicit systemic antitumor immunity against intraocular melanoma and its metastasis. J Immunother Cancer 2021, 9(1).

  217. Dayal N, Mikek CG, Hernandez D, Naclerio GA, Yin Chu EF, Carter-Cooper BA, Lapidus RG, Sintim HO. Potently inhibiting cancer cell migration with novel 3H-pyrazolo[4,3-f]quinoline boronic acid ROCK inhibitors. Eur J Med Chem. 2019;180:449–56.

    Article  CAS  PubMed  Google Scholar 

  218. Shang X, Marchioni F, Evelyn CR, Sipes N, Zhou X, Seibel W, Wortman M, Zheng Y. Small-molecule inhibitors targeting G-protein-coupled rho guanine nucleotide exchange factors. Proc Natl Acad Sci USA. 2013;110(8):3155–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Kitzing TM, Sahadevan AS, Brandt DT, Knieling H, Hannemann S, Fackler OT, Grosshans J, Grosse R. Positive feedback between Dia1, LARG, and RhoA regulates cell morphology and invasion. Genes Dev. 2007;21(12):1478–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Thompson WR, Yen SS, Uzer G, Xie Z, Sen B, Styner M, Burridge K, Rubin J. LARG GEF and ARHGAP18 orchestrate RhoA activity to control mesenchymal stem cell lineage. Bone. 2018;107:172–80.

    Article  CAS  PubMed  Google Scholar 

  221. Chiu WC, Chiang JY, Chiang FT. Small chemical compounds Y16 and Rhosin can inhibit calcium sensitization pathway in vascular smooth muscle cells of spontaneously hypertensive rats. J Formos Med Association = Taiwan Yi Zhi. 2021;120(10):1863–8.

    Article  CAS  PubMed  Google Scholar 

  222. Lorenzo-Martín LF, Rodríguez-Fdez S, Fabbiano S, Abad A, García-Macías MC, Dosil M, Cuadrado M, Robles-Valero J, Bustelo XR. Vav2 pharmaco-mimetic mice reveal the therapeutic value and caveats of the catalytic inactivation of a rho exchange factor. Oncogene. 2020;39(28):5098–111.

    Article  PubMed  PubMed Central  Google Scholar 

  223. Lorenzo-Martín LF, Menacho-Márquez M, Bustelo XR. Drug vulnerabilities and Disease Prognosis Linked to the Stem Cell-Like Gene expression program triggered by the RHO GTPase activator VAV2 in Hyperplastic keratinocytes and Head and Neck Cancer. Cancers 2020, 12(9).

  224. Zhong WB, Liang YC, Wang CY, Chang TC, Lee WS. Lovastatin suppresses invasiveness of anaplastic thyroid cancer cells by inhibiting rho geranylgeranylation and RhoA/ROCK signaling. Endocrine-related Cancer. 2005;12(3):615–29.

    Article  CAS  PubMed  Google Scholar 

  225. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.

    Article  CAS  PubMed  Google Scholar 

  226. Tnimov Z, Abankwa D, Alexandrov K. RhoGDI facilitates geranylgeranyltransferase-I-mediated RhoA prenylation. Biochem Biophys Res Commun. 2014;452(4):967–73.

    Article  CAS  PubMed  Google Scholar 

  227. Abe H, Kamai T, Hayashi K, Anzai N, Shirataki H, Mizuno T, Yamaguchi Y, Masuda A, Yuki H, Betsunoh H, et al. The rho-kinase inhibitor HA-1077 suppresses proliferation/migration and induces apoptosis of urothelial cancer cells. BMC Cancer. 2014;14:412.

    Article  PubMed  PubMed Central  Google Scholar 

  228. Nakabayashi H, Shimizu K. HA1077, a rho kinase inhibitor, suppresses glioma-induced angiogenesis by targeting the Rho-ROCK and the mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (MEK/ERK) signal pathways. Cancer Sci. 2011;102(2):393–9.

    Article  CAS  PubMed  Google Scholar 

  229. Yang X, Di J, Zhang Y, Zhang S, Lu J, Liu J, Shi W. The rho-kinase inhibitor inhibits proliferation and metastasis of small cell lung cancer. Biomed Pharmacotherapy = Biomedecine Pharmacotherapie. 2012;66(3):221–7.

    Article  CAS  PubMed  Google Scholar 

  230. Takeda H, Okada M, Suzuki S, Kuramoto K, Sakaki H, Watarai H, Sanomachi T, Seino S, Yoshioka T, Kitanaka C. Rho-Associated protein kinase (ROCK) inhibitors inhibit Survivin expression and sensitize pancreatic Cancer stem cells to Gemcitabine. Anticancer Res. 2016;36(12):6311–8.

    Article  CAS  PubMed  Google Scholar 

  231. Yang S, Kim HM. ROCK inhibition activates MCF-7 cells. PLoS ONE. 2014;9(2):e88489.

    Article  PubMed  Google Scholar 

  232. Bhandary L, Whipple RA, Vitolo MI, Charpentier MS, Boggs AE, Chakrabarti KR, Thompson KN, Martin SS. ROCK inhibition promotes microtentacles that enhance reattachment of breast cancer cells. Oncotarget. 2015;6(8):6251–66.

    Article  PubMed  Google Scholar 

  233. Tilson SG, Haley EM, Triantafillu UL, Dozier DA, Langford CP, Gillespie GY, Kim Y. ROCK inhibition facilitates in Vitro Expansion of Glioblastoma Stem-Like cells. PLoS ONE. 2015;10(7):e0132823.

    Article  PubMed  Google Scholar 

  234. Ohata H, Ishiguro T, Aihara Y, Sato A, Sakai H, Sekine S, Taniguchi H, Akasu T, Fujita S, Nakagama H, et al. Induction of the stem-like cell regulator CD44 by rho kinase inhibition contributes to the maintenance of colon cancer-initiating cells. Cancer Res. 2012;72(19):5101–10.

    Article  CAS  PubMed  Google Scholar 

  235. Sadok A, McCarthy A, Caldwell J, Collins I, Garrett MD, Yeo M, Hooper S, Sahai E, Kuemper S, Mardakheh FK, et al. Rho kinase inhibitors block melanoma cell migration and inhibit metastasis. Cancer Res. 2015;75(11):2272–84.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Editage for the manuscript language editing.

Funding

This work was supported by grants from the National Science Foundation of China (#82173283 and #82103088) and the Foundation of the committee on science and technology of Tianjin (#21JCZDJC00230, 21JCYBJC01070, 22JCZDJC00310). The funders had no roles in the design of the study, data collection, analysis, interpretation, or decision to write and publish the work.

Author information

Authors and Affiliations

Authors

Contributions

YN designed the paper, contributed to manuscript writing, and approved the manuscript before submission. MZ, and YZ collected data and performed manuscript writing. YJ and JW reviewed the literature and approved the manuscript before submission. SZ reviewed and edit the manuscript, and approved the manuscript before submission.

Corresponding author

Correspondence to Shiwu Zhang.

Ethics declarations

Ethics approval and consent to participate

This review does not contain any studies with human or animal subjects performed by any authors.

Consent for publication

All the authors have approved the manuscript and agree with submission.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ning, Y., Zheng, M., Zhang, Y. et al. RhoA-ROCK2 signaling possesses complex pathophysiological functions in cancer progression and shows promising therapeutic potential. Cancer Cell Int 24, 339 (2024). https://doi.org/10.1186/s12935-024-03519-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-024-03519-7

Keywords