Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 14.
Published in final edited form as: Biochem Soc Trans. 2021 Dec 17;49(6):2941–2955. doi: 10.1042/BST20211071

Role of Rho GTPases in stem cell regulation

Zheng Zhang 1, Ming Liu 2, Yi Zheng 1
PMCID: PMC9008577  NIHMSID: NIHMS1789964  PMID: 34854916

Abstract

The future of regenerative medicine relies on our understanding of stem cells which are essential for tissue/organ generation and regeneration to maintain and/or restore tissue homeostasis. Rho family GTPases are known regulators of a wide variety of cellular processes related to cytoskeletal dynamics, polarity and gene transcription. In the last decade, major new advances have been made in understanding the regulatory role and mechanism of Rho GTPases in self-renewal, differentiation, migration, and lineage specification in tissue-specific signaling mechanisms in various stem cell types to regulate embryonic development, adult tissue homeostasis, and tissue regeneration upon stress or damage. Importantly, implication of Rho GTPases and their upstream regulators or downstream effectors in the transformation, migration, invasion and tumorigenesis of diverse cancer stem cells highlights the potential of Rho GTPase targeting in cancer therapy. In this review, we discuss recent evidence of Rho GTPase signaling in the regulation of embryonic stem cells, multiple somatic stem cells, and cancer stem cells. We propose promising areas where Rho GTPase pathways may serve as useful targets for stem cell manipulation and related future therapies.

Introduction

Mammalian Rho family GTPases are members of the Ras superfamily including over 20 gene products which can be further subdivided into six groups based on sequence identity, domain structure and function: RhoA subfamily proteins, Rac proteins, CDC42 proteins, Rnd proteins, Rho BTB proteins and Miro proteins [1,2]. Rho family GTPases play critical roles in multiple cell processes including gene expression, cytoskeletal dynamics, survival, cell division, cell adhesion, polarity and vesicle trafficking [3,4]. Dysfunction of Rho GTPase regulated signaling can lead to multiple cancers, neurodegenerative disorders, immunological disorders, and developmental abnormalities [5]. Most Rho family members act as molecular switches that cycle between an active GTP-bound form and an inactive GDP-bound form and active Rho GTPases turn on a cascade of signaling events leading to cellular responses.

Stem cells are characterized by their ability to perpetuate themselves through self-renewal and giving rise to mature specialized cell types [6]. Based on their potency and origins, naturally occurring stem cells can be categorized into embryonic and somatic. Embryonic stem cells (ESCs) originate from inner cell mass of the blastocyst and are pluripotent with an indefinite replicative capability. Somatic or adult stem cells, on the other hand, are less totipotent with limited replicative life span than ESCs, and exist in mature tissues such as hematopoietic, neural, gastrointestinal and mesenchymal to sustain life-long tissue homeostasis. The ability of adult stem cells to divide and regenerate mature cells of a particular tissue to replace damaged tissues provides the organism with an internal repair system in response to tissue damage and stress. Tremendous efforts have been made to understand how diverse signaling pathways and regulators coordinate stem cell functions during development, homeostasis and disease/injury. The future of regenerative medicine lies in developing cell-replacement therapies to treat debilitating diseases founded on stem cell research [7].

A major focus of stem cell biology is to understand the molecular and cellular mechanisms underlying stem cell self-renewal and differentiation. In homeostasis, stem cells must strike a balance between self-renewal and differentiation, and recent studies using mouse gene targeting and small molecule inhibitors of individual Rho GTPases such as RhoA, Rac1, and Cdc42 have established that the Rho GTPase family is one of the essential regulators of embryonic and adult stem cells by controlling tissue-specific stem cell migration, niche interaction, proliferation/survival, self-renewal and differentiation (Table 1). In this review, we summarize and update recent discoveries of Rho GTPase signaling in stem cell regulation.

Table 1.

Summary of the reported roles of Rho GTPases in various stem cells

Cell type Rho GTPase Function and Mechanism Reference
Embryonic Stem Cells (ESCs) Rho/ROCK Cause dissociation-induced apoptosis of human ESCs and dramatically decrease cloning efficiency [1]
Promote colony formation and maintain them at undifferentiated state at high seeding density [2]
RhoA Mediates fibronectin-regulated mouse ESCs proliferation and migration [3]
Maintain cell–cell contacts in mouse ESCs [4]
Associated with enhanced self-renewal and reduced differentiation in ZO-1 null mouse ESCs [5]
RhoC Required for human ESCs survival and long-term growth in vitro via modulating actin microfilament organization and YAP/TAZ signaling [6]
Rac1/Cdc42 Increases mouse ESCs migration via PAK1 mediated E-cadherin complex disruption [7]
Hematopoie-tic stem cells (HSCs) RhoA Critical for multilineage hematopoiesis by regulates actomyosin signaling, cytokinesis, and programmed necrosis of the hematopoietic progenitors [8]
RhoC Regulated by RhoGAP Arhgap21, it reduces HSCs’ ability to form progenitor colonies in vitro and engraftment ability in vivo, along with a decrease in LSK cell frequency during serial bone marrow transplantation [9]
Cdc42 Elevated in aging HSCs and mediates non-canonical Wnt5A induced aging phenotypes in HSCs [10]
Control HSCs asymmetric division and fate by forming complex with Scribble/Yap1 [11]
Rac1/Rho Enhanced Rac/Rho/integrin signaling by MLL1 induction increases hematopoietic potential [12]
Neural stem cells (NSCs) RhoA/Rac1 RhoA activation and Rac1 inhibition inhibit NSCs migration by reducing actin filament polymerization [13]
RhoA Activation in hippocampal progenitors suppresses neurogenesis by regulating cellular stiffness [14]
Inhibition of RhoA leads to neurite outgrowth of NSCs and improves neuronal differentiation of NSCs [15]
Loss of RhoA impairs neural progenitor cell differentiation and cause delay during embryonic neurogenesis due to disrupted cell cycle gene expression [16]
Deletion of RhoA in midbrain and forebrain neural progenitors cause massive dysplasia of the brain due to accelerated proliferation, reduction in cell-cycle exit, and increased expression of downstream target genes of the hedgehog pathway [17]
Rac1 Promote NSC migration by regulating actin cytoskeleton [18]
Required for the normal proliferation and differentiation of SVZ progenitors and for survival of both VZ and SVZ progenitors [19]
Cdc42 Maintains NSC quiescence by inducing expression of molecules involved in NSCs identity and anchorage to the niche [20]
Rnd3/RhoE Maintain radial fiber scaffold responsible for guided neurons migration by inhibiting Notch signaling [21]
Intestinal stem cell (ISCs) RhoA RhoA activate canonical Wnt and YAP signaling, and RhoA deficiency resulted in reduced cell proliferation, increased apoptosis, and a loss of ISCs [22]
Cdc42 Required for the functioning of the Lgr5+ ISCs by activating Rab8a and its multiple effectors [23]
Loss of CDC42 caused intestinal hyperplasia and ISCs to TA cells fate switch by regulating YAP-Ereg-mTOR signaling (author’s unpublished data)
Ectopic CDC42 enhances ISCs regeneration by mediating EGF-stimulated, receptor-mediated endocytosis and sufficient to promote MAPK signaling [24]
Loss of CDC42 promotes regenerative potential of aged ISCs [25]
Rac1 Necessary and sufficient to drive ISC proliferation and regeneration in an ROS-dependent manner [26,27]
Mesenchymal stem cells (MSCs) RhoA/ROCK MSCs differentiate into myofibroblasts when RhoA/ROCK is turned on, into endothelial cells when turned off, and RhoA promotes formation of an extracellular matrix complex consisting of connective tissue growth factor and vascular endothelial growth factor [28]
Regulate fluid-flow-induced osteogenic differentiation via the regulation of Runx2, Sox9 and PPARgamma [29]
Inhibiting RhoA signaling promotes chondrogenic differentiation by increasing the expression of SOX-9 [30]
Suppression of ROCK signaling, concomitant with EGF and bFGF stimulation enhances the differentiation of bone marrow-derived MSCs into neurons and neuroglial cells [31]
Mediates the VEGF-induced differentiation of human and rat bone marrow-derived MSCs into endothelial cells. [32]
RhoA/Cdc42/Rac1 Mediates TGFbeta-induced myofibroblast differentiation of MSCs via actin polymerization [33]
Rac1/Cdc42 Regulate migration of human MSCs via E-cadherin reduction [34]
Cdc42 Elevated CDC42 activity in aged rat adipose-derived MSC leads to senescence [35]
Cancer Stem Cells (CSCs) RhoA/ROCK Promotes breast CSC formation by YAP/TAZ activity in breast CSCs [36]
Targeting Rho/ROCK Pathway is important for radiation resistance of CSCs [37]
RhoC Regulates metastatic potential of breast CSCs and tightly associated with the breast CSCs marker ALDH1 [38]
Cdc42/Rac1 Associated with cancer stem cell properties including holoclone, sphere formation, invasion [39]
Rac1 Promotes the self-renewal of breast CSCs and mediates TAZ activation by VEGF signaling [40]
Regulate migration and promote chemotherapeutic sensitivity in Leukemia stem cells via regulating the expression of several cell intrinsic cell cycle inhibitors, as well as the extrinsic molecules that mediated the interaction of Leukemia stem cells with osteoblastic niche [41]
Involved in the glioma stem-like cell maintenance and tumorigenicity via the Rac1-Pak signaling [42]
Promotes the EMT in gastric adenocarcinoma and the acquisition of a CSC state [43]
Regulates proliferation, invasion, sphere formation and lung colonization in in human NSCLA [44]
Epidermal stem cells Cdc42 Essential for epidermal stem cells and epidermal development via Cdc42-SPRR pathway, which may correlate with epidermal barrier function [45]
RhoA/Rac1 Required for epidermal stem cell migration by regulating F-actin polymerization [46]
Rac1 Indispensable for the fate of epidermal stem cells by controlling their exit from the stem cell niche [4749]
Human dental pulp stem cells Rho/ROCK Implicated in the differentiation of human dental pulp stem cells by down-regulation of Runx2 expression [50]

Rho GTPases in embryonic stem cells

ESCs are cells derived from the early embryo that can be maintained in the primitive undifferentiated state indefinitely while remaining pluripotent [8]. Rho GTPases have been implicated in multiple physiological and pathological processes in the ESC regulation (Figure 1).

Figure 1. Schematics of the recognized role of Rho GTPases in embryonic stem cells.

Figure 1.

RhoA regulates fibronectin mediated ESC migration, while Rac1 and Cdc42 are activated by laminin to enhance E-cadherin complex disruption and increase ESC migration. ROCK inhibition via Y-27632 significantly decreases ESC apoptosis while RhoA activity is associated with enhanced self-renewal and reduced differentiation. RhoC signaling is required for human ESC survival and long-term growth.

The Rho-associated kinase (ROCK) inhibitor, Y-27632, could markedly reduce dissociation-induced apoptosis of human ESCs and significantly increase cloning efficiency [9], as a ‘Rho-high/Rac-low’ state appears to initiate dissociation-induced actomyosin hyperactivation and apoptosis in the ESCs [10]. Y-27632 has also been used to improve human ESC pluripotency [11] where ROCK activation is thought to affect cellular motility [12]. More recent study found that RhoC signaling is required for human ESC survival and long-term growth in vitro by modulating actin microfilament organization and sustaining the nuclear function of the transcriptional cofactors YAP and TAZ [13]. Furthermore, it was reported that ABR, a modulator of Rho GTPase activities, is an upstream factor controlling the survival vs. death decision of dissociated human ESCs [14].

Murine ESC adhesion to fibronectin stimulated cell proliferation and migration in a RhoA dependent manner [15,16]. Functional analysis of the promoters of the RhoA and the RhoB genes in mouse ESCs showed significantly higher RhoA mRNA levels than RhoB, and expression of both genes was inhibited by their own protein products [17]. In addition, RhoA is also required in early embryogenesis for the maintenance of intercellular junctions in mouse ESCs [18]. Separately, Rac1/Cdc42 can be activated by laminin, the first extracellular matrix component expressed in the developing mammalian embryo, to enhance E-cadherin complex disruption leading to increased ESCs migration [19].

The role of Rho GTPases in the differentiation of ESCs is relatively under studied. Due to the similarity of ESCs and induced pluripotent stem cells (iPSCs) which is undergoing fast-evolving studies, understanding how Rho GTPases regulate the controlled differentiation of iPSCs towards different tissue specific progenitor lineages will help gain insights into ESC differentiation mechanisms in the future.

Rho GTPases in hematopoietic stem cells

While adult stem cells from a variety of organs have the potential to be useful for therapeutic applications in the future, the hematopoietic stem cells (HSCs) have already been extensively used for clinical purposes, mainly by bone-marrow or cord blood-related transplantation. HSCs and hematopoietic progenitor cell (HPCs) in the bone marrow or cord blood initiate the production of multilineage hematopoietic cells via two critical features of HSCs: indefinite self-renewal and multilineage differentiation potential. Rho GTPases are required for most aspects of HSC functions including lineage determining gene transcription, cell survival, cell cycle progression and homing and migration. Alterations of HSC function through Rho GTPases contribute to various human diseases including leukemogenesis, bone marrow failure syndromes, immunodeficiencies, and blood aging phenotypes [1] (Figure 2). The function of Rho GTPases especially Rac1 and Rac2 in the hematopoietic system has been well studied in HSCs and mature hematopoietic cells, and earlier work has been summarized [20]. Research focusing on their role in HSCs have revealed the essential contributions of RhoA, Rac1/2, Cdc42 and other Rho GTPases/regulators in HSC adhesion, migration, bone marrow retention, proliferation, survival, senescence and oncogenic transformation [1,21].

Figure 2. Schematics of the role of Rho GTPases in hematopoietic stem cells.

Figure 2.

RhoA regulates actomyosin signaling, cytokinesis, and programmed necrosis of the hematopoietic progenitors. Cdc42 is a master regulator of HSC engraftment, quiescence, niche interaction, and aging associated loss of polarity phenotypes, while Rac1/Rac2 play critical roles in the HSC engraftment and migration.

Earlier studies of the role of RhoA in HSC regulation utilized mouse models expressing dominant negative RhoA (RhoAN19) and showed that partial inhibition of RhoA caused an enhanced HSC self-renewal activity and long-term engraftment capability [22]. Whereas loss of p190-B Rho GTPase activating protein, a negative regulator of RhoA, induced elevated RhoA activity and the associated increased HSC self-renewal activity and long-term engraftment [23]. Subsequent RhoA conditional knockout in a mouse model demonstrated directly that RhoA deficiency in HSCs caused multilineage hematopoietic failure with a blockage of hematopoiesis at the multipotent progenitor stage [24]. RhoA-null HSCs still possess long-term engraftment potential but are incapable of producing multipotent progenitors and differentiated blood cells, as RhoA regulates actomyosin signaling, cytokinesis, and programmed necrosis of the hematopoietic progenitors [24]. Later studies in human patients found that vasculopathy-associated hyperangiotensinemia induces a mobilization of HSCs and progenitors through endothelial AT2R and RhoA signaling [25].

Studies of the function of other Rho GTPases have found that CDC42 is a master regulator of HSC engraftment, quiescence, niche interaction, and aging associated loss of polarity phenotypes [26,27]. Expression of Wnt5a and Cdc42 are elevated in aged primitive hematopoietic cells and Wnt5a activated Cdc42 is involved in the induction of ageing-like myeloid skewage and HSC engraftment defects of HSCs [28]. The recently identified Cdc42 activity-specific inhibitor, CASIN, could inhibit intracellular Cdc42 activity transiently and cause mouse HSCs and progenitor cell egress from bone marrow by suppressing actin polymerization, adhesion, and directional migration of HSCs, which is useful to study biological and pathological roles of Cdc42 in HSCs as well as other tissue cells [29]. More recent work have shown that the cell planar polarity molecule Scribble and the Hippo pathway component Yap1 coordinate to control cytoplasmic Cdc42 activity and HSC fate in vivo [30]. On the other hand, increased RhoC activity in HSCs by loss of Arhgap21, a member of the Rho GAP family, led to impaired ability to form progenitor colonies in vitro and decreased engraftment ability in vivo, along with a decrease in multiple potential progenitor cell frequency in serial bone marrow transplantation and reduced erythroid commitment and differentiation [31]. Studies in zebrafish and iPSCs indicate that members of the Rho GTPase family act upstream of YAP to regulate HSCs and progenitor cell production [32]. Induction of MLL1, a gene critical for development and maintenance of HSCs, in mouse ESCs enhances Rac1/RhoA/integrin signaling and selectively affects c-Kit+/CD41+ hematopoietic progenitor function and increases the ESC differentiated HPC hematopoietic potential [33].

The role of Rho GTPases in HSCs is one of the most studied among stem cell subtypes. RhoA, Rac and Cdc42 all have been demonstrated to have essential roles in multiple aspects of HSC function. Ongoing studies of Rho GTPases and their regulators/inhibitors in HSCs and hematological diseases are moving toward translation in HSC transplantation, HSC aging, and myeloid dysplasia/transformation arenas.

Rho GTPases in neural stem cells

Neural stem cells (NSCs) reside in the ventricular and subventricular zones (VZ and SVZ) and give rise to the highly elongated radial glial progenitors as the central nervous system (CNS) matures during development [34,35] (Figure 3). NSCs play key roles in the developing CNS, including generating neurons and macroglial cells and guiding the migration of newborn neurons to their proper destinations [36]. In adult, NSCs are largely quiescent during homeostasis, and upon CNS injury, NSCs begin proliferating and differentiating to three major CNS cell types — neurons, astrocytes, and oligodendrocytes — while migrating towards the lesions to repair injury [37]. The molecular mechanisms that trigger the activation of NSCs for neural tissue repair were unclear until a recent study found that NSC quiescence is maintained by Cdc42 which is in turn regulated by non-canonical Wnt signaling [38]. Cdc42 induces the expression of molecules involved in NSC identity maintenance and anchor the NSCs to the niche, while during demyelination injuries, down-regulation of Cdc42 activity is required for the activation and lineage progression of quiescent NSCs for tissue repair [38].

Figure 3. Schematics of the key roles of Rho GTPases in neural stem cells.

Figure 3.

Cdc42 maintains NSC quiescence, and down-regulation of Cdc42 activity results in the activation of quiescent NSCs for tissue repair. NSC migration relies heavily on Rho GTPase (Rac1, RhoA and Rnd3/RhoE)-controlled actin and microtubule cytoskeleton, whereas RhoA has diverse roles during neurogenesis. The blue and brown tracts indicate the blood vessels.

NSC migration relies heavily on the regulation of actin and microtubule cytoskeleton, and Rho GTPases are critical regulators of key steps during NSC migration [39,40]. Fasudil, a ROCK inhibitor, drives mobilization of adult neural stem cells after hypoxia/reoxygenation injury in mice [41], and overexpression of Rho-GDIγ (RhoGDI-3/ARHGDIG), mainly a down-regulator of Rho GTPases while also having a positive role as chaperones in certain cell-context, maintains NSCs in the stem cell state, prevents NSC migration and inhibit of Rac1 expression [42]. A recent study found that the maintenance of radial fiber scaffold responsible for guided neurons migration, and consequently axon pathfinding, are dependent on an atypical Rho-GTPase, Rnd3/RhoE and its binding partner ARHGAP35/p190A, a RHO GTPase-activating protein [36], and genetic deletion of Rnd3 in NSCs promotes their proliferation via up-regulation of Notch signaling [43]. In cultured primary NSCs, RhoA activation and Rac1 inhibition caused by the loss of actin alpha 2 result in reduced actin filament polymerization and an inhibition of NSC migration [37], suggesting that the regulation of actin and microtubule cytoskeleton by Rho GTPases is important for NSC migration.

Self-renewal and differentiation of NSCs depend not only on the intrinsic biochemical and genetic factors but also on the mechanical cues in their microenvironment such as extracellular matrix (ECM) stiffness [44]. ECM-derived mechanical signals act through Rho GTPases to activate the cellular contractility machinery and regulate NSC lineage commitment, as NSCs cultured on stiff ECMs showed elevated RhoA and Cdc42 activities and suppressed neurogenesis, whereas RhoA activation in hippocampal progenitors also suppresses neurogenesis [45]. During in vitro culturing of human NSCs, it is found that YAP-mediated mechano-sensing regulates neurogenesis and lineage commitment of the NSCs and the intracellular localization of YAP is regulated by RhoA activity [46]. A later study found that RhoA inhibition can also affect neuritogenesis and differentiation of NSCs by promoting neuronal differentiation and neurite outgrowth in NSCs isolated from mouse SVZ [47]. A recent study identified a Rho GTPase-activating protein (RhoGAP), α2-chimaerin, in regulating the transition of NSCs into intermediate progenitor cells, and conditional knockout of α2-chimaerin resulted in a loss of NSC population by premature differentiation with impaired neuron production [48]. On the contrary, suppression of RhoA activity by a dominant-negative RhoA mutant impaired neural progenitor cell differentiation and caused a delay of embryonic neurogenesis [49], suggesting that RhoA has diverse roles at different developmental stages. Interestingly, spontaneous neural differentiation from mouse ESCs can be induced by Noggin, and RhoA activity in the ESCs prevents neural differentiation by inhibiting Noggin transcription and limiting Noggin secretion [50]. Furthermore, ROCK inhibitor Y-27632 treatment of ESCs promotes the differentiation of the cells into neurons [51].

More detailed analyses of various populations of neural stem/progenitor cells have seen more selective roles of various Rho GTPases in the regulation of self-renewal and differentiation. Conditional deletion of RhoA in midbrain and forebrain neural progenitors disrupted apical adhesion junctions and caused a massive dysplasia of the brain due to accelerated proliferation, reduction in cell-cycle exit, and increased expression of downstream target genes of the hedgehog pathway, resulting in an expansion of neural progenitor cells and exencephaly-like protrusions [52]. Conditional deletion of Cdc42 at different developmental stages of neurogenesis in mouse telencephalon also caused decay of adhesion junctions leading to a loss of the neuroepithelial polarity and failure of apically directed interkinetic nuclear migration [53]. On the other hand, Cdc42 deficiency in the developing forebrain stem cells disrupted apical-basal polarity and led to disorganized and expanded neural progenitors throughout the entire neuroepithelium [54].

Thus, Rho GTPases are important for the activation, migration and differentiation of NSCs and neural progenitors (Figure 3). Due to their important function in regulating polarity and cell adhesions, it is not surprising that individual Rho GTPases are critical for migration-related functions of NSCs in injury recovery and posits them as valuable candidate targets in the treatments of neuronal injuries and neuronal degenerative diseases.

Rho GTPases in intestinal stem cells

Intestinal epithelium is one of the most vigorously self-renewing tissues in adult mammals; the entire lining of the human gut replenishes approximately every 7 days [55]. The function of several Rho GTPases in intestinal stem cell regulation (ISCs) has become more clear recently (Figure 4). Genetic deletion of Cdc42 in mice results in massive proliferation, disrupted epithelium morphogenesis, impaired intestinal epithelium polarity, and defective Paneth cell localization [56]. Another Cdc42 knockout study found that Cdc42 deficiency perturbs crypt homeostasis and reduced the function of the Lgr5+ ISCs [57]. An ISC-specific deletion of Cdc42 in mice found that Cdc42 controls apical/basal lateral polarity which regulates ISC to transit amplifying (TA) cell transition and proliferation at the expense of ISC population via a canonical Hippo signaling of YAP/TAZ and the consequent Ereg mediated mTOR pathway (author’s unpublished data), positioning Cdc42 regulated epithelial polarity at the center of ISC/TA cell proliferation and cell fate control. Recent studies have also found that pharmacological suppression of elevated Cdc42 activity restores the regenerative potential of aged ISCs [58], and extends the general lifespan of mice [59].

Figure 4. Schematics of the role of Rho GTPases in intestinal stem cells.

Figure 4.

Rac1/RhoA/Cdc42 regulate ISC proliferation and regeneration via different downstream effectors. RhoA and Cdc42 are required for intestinal epithelial polarity but distinct downstream signaling through the Hippo signaling effecting on ISC proliferation. Cdc42 uniquely contributes to the ageing effect of ISCs and ISC into TA cell transition. TA: Transit amplifying cells; PC; Paneth cells; ISC: Intestinal stem cells; GB: Goblet cells; EE: Enteroendocrine cells; TC: Tuft cells.

Aside from Cdc42, RhoA GTPase also controls epithelial polarity as well as YAP-mediated Ereg signaling in ISCs as RhoA deficiency results in a reduced proliferation, increased apoptosis, and loss of ISCs that mimic effects of radiation damage [60]. On the other hand, Rac1 works downstream of canonical Wnt signaling and is necessary and sufficient to drive ISC proliferation and regeneration in an ROS-dependent manner [61,62]. Recent studies revealed further that the Wnt target Lgr5 primarily functions via the IQGAP1-Rac1 pathway to strengthen cell-cell adhesion in ISCs [63]. It remains to be seen how the interactions of each Rho GTPase with other known regulators of ISCs such as Wnt, Hippo, and Notch pathways are integrated together in the ISC maintenance, particularly after a stress in the intestine.

Rho GTPases in other somatic stem cells

While the available literature remains relatively sparse, Rho GTPases have also been implicated in the regulation of other somatic stem cell types including epidermal, teeth, cardiac and mesenchymal stem cells. In vivo study showed that Cdc42 deficiency leads to the loss of epidermal stem cells [64], whereas Rac1 and RhoA, but not Cdc42, are required for epidermal stem cell migration [65]. Furthermore, both in vivo and in vitro studies have found that loss of Rac1 causes epidermal stem cell depletion [6668]. The ROCK inhibitor Y-27632 promotes proliferation of stem cell from human exfoliated deciduous teeth [69], and the Rho/ROCK signaling is implicated in the differentiation of human dental pulp stem cells [70]. In the growing mouse incisor, Cdc42 was found to activate YAP which drives the proliferation of transit-amplifying cells and affects stem cell-based tissue renewal in mice [71]. Additionally, Y-27632 significantly increased cell survival rate, adhesion, and migration in cardiac stem cells [72].

It has been shown that TGFβ-activated RhoA/ROCK signaling functions as a molecular switch regarding the fate of mesenchymal stem cells (MSCs) in arterial repair/remodeling after injury, and MSCs can differentiate into myofibroblasts when RhoA/ROCK is turned on but to endothelial cells when turned off [73]. In bone marrow, RhoA and its effector ROCKII regulate fluid-flow-induced osteogenic differentiation [74], while RhoA GEFs and GAPs work through RhoA to influence MSC fate [75]. The function of Rho GTPases in skeletal myogenesis was summarized previously [76]. Interestingly, Vav1, a Rho/Rac GEF highly expressed in MSCs, regulates MSC differentiation decision between adipocyte and chondrocyte [77]. Suppression of ROCK signaling enhances the efficacy of bone marrow-derived MSC differentiation into neurons and neuroglial cells [78], and pharmacologic experiments found that elevated Cdc42 activity in aged rat adipose-derived MSCs leads to senescence and reduced differentiation potential while inhibition of Cdc42 activity can, at least partially, rejuvenate aged MSCs [79].

Rho GTPases in cancer stem cells

The cancer stem cell (CSC) theory proposes that a tumor is hierarchical in cellular components and there is a subpopulation of tumor cells with enhanced self-renewal and tumor seeding activities upon experimental implantation [80] (Figure 5). In many myeloid leukemia and solid tumor types including acute myeloid leukemia, glioblastoma, breast cancer, colorectal cancer and skin squamous-cell cancers [81], CSCs are active architects of their microenvironment to maintain a supportive niche [82]. In many aforementioned tumor types, only a small subset of the cancer cells contains the capability of tumor invasiveness, therapy resistance and tumor initiation, positing CSCs in the central scheme of cancer biology with great clinical potentials [81,83].

Figure 5. Schematics of the function of individual Rho GTPases in select cancer stem cells.

Figure 5.

Rho GTPases contribute to the initiation and progression of multiple types of tumors, and play major roles of the metastatic process. ROCK and Rac1 are implicated in the chemo/radiation therapy resistance, and together with other pro-oncogenic Rho GTPases, are emerging as novel therapeutic targets.

In human breast cancer, the CSC marker ALDH1 is tightly correlated with RhoC expression, and in vitro analysis and xenograft studies further suggest that RhoC regulates ALDH1 and endows a metastatic potential of breast CSCs [84]. Recent studies found that a STARD13-correlated ceRNA network, which is inhibitory to breast CSCs, is negatively correlated with RhoA-ROCK signaling mediated YAP/TAZ activity, suggesting that inactivating RhoA GTPase/F-actin pathway might be a useful approach to suppress YAP/TAZ activity and inhibit breast CSC formation [85]. Accumulating evidence have shown that YAP/TAZ of the hippo pathway are essential for the function of CSCs and other aspects of aggressive tumor behavior [86], and Rac1 is critical in TAZ activation by VEGF signaling which promotes the self-renewal of breast CSCs [87]. Using selective pharmacologic inhibitors, it is found that in gastric CSC-like cells, activation of PAR1 inhibits Lats1/2 kinase and activates YAP/TAZ activity via a ROCK-independent Rho GTPase pathway [88], consistent with a key role of Rho family GTPases in promoting YAP/TAZ activation observed in endothelial cells [89].

CSCs derived from melanoma and breast cancer cell lines exhibit increased contractility as well as ECM remodeling capacity, and more importantly, a blockade of ROCK signaling completely abolishes the contractility and collagen degradation capacity of both CSCs and non-CSCs, suggesting a potential of pharmacological targeting of ROCK pathway in CSCs and overall tumor population in the treatment of metastasis of these cancers [90]. Interestingly, forced expression of miR-141 in prostate CSCs and progenitor cells directly inhibits Rho GTPase family members/signaling partners Cdc42, CDC42EP3 (BORG2/CEP3), Rac1 and ARPC5 (a component of the Arp2/3 complex), causing reduced CSC-related activities including holoclone, sphere formation, invasion, as well as tumor regeneration and metastasis [91]. On the other hand, RhoA knockout fibroblasts lose their normal tumor-inhibitory capacity and gain the tumor stem cell-promoting properties including induced cancer cell migration and proliferation in vitro and tumor growth in vivo [92], whereas ROCK inhibitor Y-27632 and fasudil can sensitize pancreatic CSCs to chemotherapy response to Gemcitabine [93]. There is also evidence that the Rho/ROCK Pathway are involved in radio-resistance and targeting Rho/ROCK Pathway offers a potential solution to overcoming radiation resistance of CSCs [94].

Inactivation of Rac1 causes an impaired migration and enhanced chemotherapeutic sensitivity in leukemia stem cells [95]. In human glioma cell lines, Rac1 is involved in the glioma stem-like cell maintenance and tumorigenicity [96]. Rac1 activity is also significantly higher in spheroid-forming or CD44+ gastric adenocarcinoma CSCs, and a cell line study using a xenograft model indicated that Rac1 promotes the epithelial-to-mesenchymal transition (EMT) in gastric adenocarcinoma and the acquisition of a CSC state [97]. Furthermore, knockdown of Rac1 in human non-small cell lung adenocarcinoma (NSCLA) cells decreases proliferation, invasion, sphere formation and lung colonization [98].

While solid tumors require Rho GTPases for migration, dissemination and metastasis, hematological malignancies depend on Rho GTPases for survival and proliferation. In acute myeloid leukemia, elevated Cdc42 activity was associated with a polarized transformation of HSPCs to acute myeloid leukemia (AML) with altered cell division symmetry whereas elevated Rac1/Rac2 activity is crucial for Bcl-2 mediated leukemia initiating cell survival; a knockdown or small molecule targeting of Cdc42 or Rac in primary murine or human AML cells suppressed leukemia progression in xenograft models [99,100], implicating Cdc42 and Rac as potential targets for inhibiting leukemia stem cell. In solid tumors, hyperactive Rac1 and Cdc42 are implicated in EMT, cell-cycle progression, migration/invasion, tumor growth, angiogenesis, and oncogenic transformation, and an up-to-date review of Rac1 and Cdc42-targeting small molecules has been summarized elsewhere [101]. On the other hand, the RhoA gene is often deleted or mutated rather than amplified in many cancer types, suggesting a tumor suppressor role in defined cancer types [2]. However, RhoA inhibitors still have clinic potential for certain cancers where RhoA is activated. For example, a small molecule Y16 which selectively suppressed RhoA GEF signaling was able to inhibit mammary sphere formation in MCF7 breast cancer cells [102], and more recently, JK-136 and JK-139, two of the anti-RhoA hydrazide derivatives, were found to inhibit gastric cancer activities in mice [103]. So far, the majority of Rho GTPase inhibitors remain at the pre-clinical testing stage, but pharmaceutical development and research of Rho GTPase inhibitors will continue to hold promise as a therapeutic strategy.

Last but not the least, most of our understanding of Rho GTPase function in CSCs were obtained using cancer cells in vitro or in xenografts in immunocompromised mice which lack consideration of the tumor microenvironment. The development of improved preclinical models such as 3D co-culture, organoids and patient-derived xenografts in humanized animal models will increase our understanding for the role of Rho GTPases in CSCs.

Overall, Rho GTPases play diverse roles (either pro-oncogenic or tumor-suppressive) in cancer stem cells depending on the tissue of origin, progression stage, and degree of differentiation (Figure 5), presenting a challenge for rational design of anti-cancer therapies based on targeting Rho GTPase signaling.

Conclusion

Rho family GTPases are known to regulate diverse cellular responses to morphogens, adhesion molecules, growth factors/cytokines, and chemokines involved in stem cell proliferation, polarity and cytoskeletal dynamics [104]. During the last decade, chemical inhibitors and mouse genetics have allowed rapid discoveries of the essential roles of individual Rho GTPase family members in self-renewal, differentiation, migration, survival and/or niche interaction of embryonic and various somatic stem cells (Table 1). Related studies of the large arrays of Rho GTPase regulators/effectors in stem cell regulation, which are not extensively covered here due to page limit, also add to our ever-growing knowledge of Rho GTPase mediated tissue specific signaling in stem cell subtypes presents new opportunities for stem cell manipulation and therapeutics. Understanding how Rho GTPase signaling is controlled in vivo remains a challenge and important future directions include: in-depth mechanistic investigation of the interaction between Rho GTPase regulated polarity/adhesion and other intracellular signaling pathways elicited from diverse stem cell microenvironments in temporal/spatial regulation of subtypes of stem cells, and the discovery of novel therapeutic opportunities using selective pharmaceutical activation/inhibition of Rho GTPases in physiologic or pathologic settings. Such further studies of Rho GTPase function will shine light on the basic mechanisms of stem cell biology and their translational potential in stem cell-related diseases.

Perspectives.

  • Importance of the field: Stem cell derived tissue/organ development and regeneration provide great potentials for the future of regenerative medicine. A comprehensive understanding of regulatory mechanisms in embryonic/adult stem cells as well as in cancer stem cell is crucial for stem cell biology translation to future therapies. Rho family GTPases are important regulators for diverse cell biological functions, and their role in stem cell function and as potential therapeutic targets have been actively investigated in the past decade, yielding important insights into tissue specific stem cell regulatory mechanisms.

  • Current thinking: Rho family GTPases play critical regulatory roles in multiple intracellular signaling functions of various stem cells to regulate embryonic development, adult tissue homeostasis, and tissue regeneration upon stress. In addition, Rho GTPases and their upstream regulators or downstream effectors are also involved in the transformation, migration, invasion and tumorigenesis of cancer stem cells, and recognized to be potential targets for cancer therapy.

  • Future directions: Future in-depth studies into the mechanisms on how Rho GTPase-regulated polarity/adhesion may coordinate with intracellular signaling pathways for the temporal/spatial regulation of stem cells and the discovery of novel therapeutic approaches using selective pharmaceutical activation/inhibition of Rho GTPases, will shine a light on the mechanisms of stem cell biology and reveal their translational potential in stem cell-related diseases.

Funding

NIH R01CA204895, R01AG063967, R01 HL147536, U54 DK126108 and NIH P30 DK078392.

Abbreviations

AML

acute myeloid leukemia

CNS

central nervous system

CSC

cancer stem cell

ECM

extracellular matrix

EMT

epithelial-to-mesenchymal transition

ESCs

embryonic stem cells

HSCs

hematopoietic stem cells

ISC

intestinal stem cell

MSCs

mesenchymal stem cells

NSCLA

non-small cell lung adenocarcinoma

TA

transit amplifying

Footnotes

Competing Interests

The authors declare that there are no competing interests associated with the manuscript.

References

  • 1.Cancelas JA and Williams DA (2009) Rho GTPases in hematopoietic stem cell functions. Curr. Opin. Hematol 16, 249–254 10.1097/M0H.0b013e32832c4b80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Svensmark JH and Brakebusch C (2019) Rho GTPases in cancer: friend or foe? Oncogene 38, 7447–7456 10.1038/s41388-019-0963-7 [DOI] [PubMed] [Google Scholar]
  • 3.Guo F and Zheng Y (2004) Rho family GTPases cooperate with p53 deletion to promote primary mouse embryonic fibroblast cell invasion. Oncogene 23, 5577–5585 10.1038/sj.onc.1207752 [DOI] [PubMed] [Google Scholar]
  • 4.Hanna S and El-Sibai M (2013) Signaling networks of Rho GTPases in cell motility. Cell Signal. 25, 1955–1961 10.1016/j.cellsig.2013.04.009 [DOI] [PubMed] [Google Scholar]
  • 5.Hall A (2012) Rho family GTPases. Biochem. Soc. Trans 40, 1378–1382 10.1042/BST20120103 [DOI] [PubMed] [Google Scholar]
  • 6.Zakrzewski W, Dobrzynski M, Szymonowicz M and Rybak Z (2019) Stem cells: past, present, and future. Stem Cell Res. Ther 10, 68 10.1186/s13287-019-1165-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Blau HM and Daley GQ (2019) Stem cells in the treatment of disease. N. Engl. J. Med 381, 890 10.1056/NEJMx190025 [DOI] [PubMed] [Google Scholar]
  • 8.Ilic D and Ogilvie C (2017) Concise review: human embryonic stem cells-what have we done? what are we doing? where are we going? Stem Cells 35, 17–25 10.1002/stem.2450 [DOI] [PubMed] [Google Scholar]
  • 9.Watanabe K, Ueno M, Kamiya D, Nishiyama A, Matsumura M, Wataya T et al. (2007) A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat. Biotechnol 25, 681–686 10.1038/nbt1310 [DOI] [PubMed] [Google Scholar]
  • 10.Ohgushi M, Matsumura M, Eiraku M, Murakami K, Aramaki T, Nishiyama A et al. (2010) Molecular pathway and cell state responsible for dissociation-induced apoptosis in human pluripotent stem cells. Cell Stem Cell 7, 225–239 10.1016/j.stem.2010.06.018 [DOI] [PubMed] [Google Scholar]
  • 11.Sivasubramaniyan K, Pal R, Totey S, Bhat VS and Totey S (2010) Rho kinase inhibitor y27632 alters the balance between pluripotency and early differentiation events in human embryonic stem cells. Curr. Stem Cell Res. Ther 5, 2–12 10.2174/157488810790442769 [DOI] [PubMed] [Google Scholar]
  • 12.Li L, Wang BH, Wang S, Moalim-Nour L, Mohib K, Lohnes D et al. (2010) Individual cell movement, asymmetric colony expansion, rho-associated kinase, and E-cadherin impact the clonogenicity of human embryonic stem cells. Biophys. J 98, 2442–2451 10.1016/!.bpj.2010.02.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ohgushi M, Minaguchi M and Sasai Y (2015) Rho-signaling-directed YAP/TAZ activity underlies the long-term survival and expansion of human embryonic stem cells. Cell Stem Cell 17, 448–461 10.1016/j.stem.2015.07.009 [DOI] [PubMed] [Google Scholar]
  • 14.Ohgushi M, Minaguchi M, Eiraku M and Sasai Y (2017) A RHO small GTPase regulator ABR secures mitotic fidelity in human embryonic stem cells. Stem Cell Rep. 9, 58–66 10.1016/j.stemcr.2017.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Park JH, Ryu JM and Han HJ (2011) Involvement of caveolin-1 in fibronectin-induced mouse embryonic stem cell proliferation: role of FAK, rhoA, PI3K/Akt, and ERK 1/2 pathways. J. Cell Physiol 226, 267–275 10.1002/jcp.22338 [DOI] [PubMed] [Google Scholar]
  • 16.Park JH, Ryu JM, Yun SP, Kim MO and Han HJ (2012) Fibronectin stimulates migration through lipid raft dependent NHE-1 activation in mouse embryonic stem cells: involvement of RhoA, Ca2+/CaM, and ERK. Biochim. Biophys. Acta 1820, 1618–1627 10.1016/j.bbagen.2012.05.013 [DOI] [PubMed] [Google Scholar]
  • 17.Nomikou E, Stournaras C and Kardassis D (2017) Functional analysis of the promoters of the small GTPases RhoA and RhoB in embryonic stem cells. Biochem. Biophys. Res. Commun 491, 754–759 10.1016/j.bbrc.2017.07.114 [DOI] [PubMed] [Google Scholar]
  • 18.Harb N, Archer TK and Sato N (2008) The Rho-Rock-Myosin signaling axis determines cell-cell integrity of self-renewing pluripotent stem cells. PLoS ONE 3, e3001 10.1371/iournal.pone.0003001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Suh HN and Han HJ (2010) Laminin regulates mouse embryonic stem cell migration: involvement of Epac1/Rap1 and Rac1/cdc42. Am. J. Physiol. Cell Physiol 298, C1159–C1169 10.1152/ajpcell.00496.2009 [DOI] [PubMed] [Google Scholar]
  • 20.Mulloy JC, Cancelas JA, Filippi MD, Kalfa TA, Guo F and Zheng Y (2010) Rho GTPases in hematopoiesis and hemopathies. Blood 115, 936–947 10.1182/blood-2009-09-198127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nayak RC, Chang KH, Vaitinadin NS and Cancelas JA (2013) Rho GTPases control specific cytoskeleton-dependent functions of hematopoietic stem cells. Immunol. Rev 256, 255–268 10.1111/imr.12119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ghiaur G, Lee A, Bailey J, Cancelas JA, Zheng Y and Williams DA (2006) Inhibition of RhoA GTPase activity enhances hematopoietic stem and progenitor cell proliferation and engraftment. Blood 108, 2087–2094 10.1182/blood-2006-02-001560 [DOI] [PubMed] [Google Scholar]
  • 23.Xu H, Eleswarapu S, Geiger H, Szczur K, Daria D, Zheng Y et al. (2009) Loss of the Rho GTPase activating protein p190-B enhances hematopoietic stem cell engraftment potential. Blood 114, 3557–3566 10.1182/blood-2009-02-205815 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhou X, Florian MC, Arumugam P, Chen X, Cancelas JA, Lang R et al. (2013) Rhoa GTPase controls cytokinesis and programmed necrosis of hematopoietic progenitors. J. Exp. Med 210, 2371–2385 10.1084/jem.20122348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chang KH, Nayak RC, Roy S, Perumbeti A, Wellendorf AM, Bezold KY et al. (2015) Vasculopathy-associated hyperangiotensinemia mobilizes haematopoietic stem cells/progenitors through endothelial AT(2)R and cytoskeletal dysregulation. Nat. Commun 6, 5914 10.1038/ncomms6914 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Florian MC, Dorr K, Niebel A, Daria D, Schrezenmeier H, Rojewski M et al. (2012) Cdc42 activity regulates hematopoietic stem cell aging and rejuvenation. Cell Stem Cell 10, 520–530 10.1016/j.stem.2012.04.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Yang L, Wang L, Geiger H, Cancelas JA, Mo J and Zheng Y (2007) Rho GTPase Cdc42 coordinates hematopoietic stem cell quiescence and niche interaction in the bone marrow. Proc. Natl Acad. Sci. U.S.A 104, 5091–5096 10.1073/pnas.0610819104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Florian MC, Nattamai KJ, Dorr K, Marka G, Uberle B, Vas V et al. (2013) A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature 503, 392–396 10.1038/nature12631 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Liu W, Du W, Shang X, Wang L, Evelyn C, Florian MC et al. (2019) Rational identification of a Cdc42 inhibitor presents a new regimen for long-term hematopoietic stem cell mobilization. Leukemia 33, 749–761 10.1038/s41375-018-0251-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Althoff MJ, Nayak RC, Hegde S, Wellendorf AM, Bohan B, Filippi MD et al. (2020) Yap1-Scribble polarization is required for hematopoietic stem cell division and fate. Blood 136, 1824–1836 10.1182/blood.2019004113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Xavier-Ferrucio J, Ricon L, Vieira K, Longhini AL, Lazarini M, Bigarella CL et al. (2018) Hematopoietic defects in response to reduced Arhgap21. Stem Cell Res. 26,17–27 10.1016/j.scr.2017.11.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lundin V, Sugden WW, Theodore LN, Sousa PM, Han A, Chou S et al. (2020) YAP regulates hematopoietic stem cell formation in response to the biomechanical forces of blood flow. Dev. Cell 52, 446–60 e5 10.1016/j.devcel.2020.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yang W, Trahan GD, Howell ED, Speck NA, Jones KL, Gillen AE et al. (2020) Enhancing hematopoiesis from murine embryonic stem cells through MLL1-Induced activation of a Rac/Rho/Integrin signaling axis. Stem Cell Rep. 14, 285–299 10.1016/j.stemcr.2019.12.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Taverna E, Gotz M and Huttner WB (2014) The cell biology of neurogenesis: toward an understanding of the development and evolution of the neocortex. Annu Rev Cell Dev. Biol 30, 465–502 10.1146/annurev-cellbio-101011-155801 [DOI] [PubMed] [Google Scholar]
  • 35.Breunig JJ, Haydar TF and Rakic P (2011) Neural stem cells: historical perspective and future prospects. Neuron 70, 614–625 10.1016/j.neuron.2011.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kaur N, Han W, Li Z, Madrigal MP, Shim S, Pochareddy S et al. (2020) Neural stem cells direct axon guidance via their radial fiber scaffold. Neuron 107, 1197–211 e9 10.1016/j.neuron.2020.06.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhang J, Jiang X, Zhang C, Zhong J, Fang X, Li H et al. (2020) Actin alpha 2 (ACTA2) downregulation inhibits neural stem cell migration through Rho GTPase activation. Stem Cells Int. 2020, 4764012 10.1155/2020/4764012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chavali M, Klingener M, Kokkosis AG, Garkun Y, Felong S, Maffei A et al. (2018) Non-canonical Wnt signaling regulates neural stem cell quiescence during homeostasis and after demyelination. Nat. Commun 9, 36 10.1038/s41467-017-02440-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Govek EE, Hatten ME and Van Aelst L (2011) The role of Rho GTPase proteins in CNS neuronal migration. Dev. Neurobiol 71, 528–553 10.1002/dneu.20850 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Numano F, Inoue A, Enomoto M, Shinomiya K, Okawa A and Okabe S (2009) Critical involvement of Rho GTPase activity in the efficient transplantation of neural stem cells into the injured spinal cord. Mol. Brain 2, 37 10.1186/1756-6606-2-37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ding J, Li QY, Yu JZ, Wang X, Sun CH, Lu CZ et al. (2010) Fasudil, a Rho kinase inhibitor, drives mobilization of adult neural stem cells after hypoxia/reoxygenation injury in mice. Mol. Cell. Neumsci 43, 201–208 10.1016/j.mcn.2009.11.001 [DOI] [PubMed] [Google Scholar]
  • 42.Wang J, Li X, Cheng H, Wang K, Lu W and Wen T (2013) Overexpression of Rho-GDP-dissociation inhibitor-gamma inhibits migration of neural stem cells. J. Neurosci. Res 91, 1394–1401 10.1002/jnr.23261 [DOI] [PubMed] [Google Scholar]
  • 43.Dong H, Lin X, Li Y, Hu R, Xu Y, Guo X et al. (2017) Genetic deletion of Rnd3 in neural stem cells promotes proliferation via upregulation of notch signaling. Oncotarget 8, 91112–91122 10.18632/oncotarget.20247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Saha K, Keung AJ, Irwin EF, Li Y, Little L, Schaffer DV et al. (2008) Substrate modulus directs neural stem cell behavior. Biophys. J 95, 4426–4438 10.1529/biophysj.108.132217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Keung AJ, de Juan-Pardo EM, Schaffer DV and Kumar S (2011) Rho GTPases mediate the mechanosensitive lineage commitment of neural stem cells. Stem Cells 29, 1886–1897 10.1002/stem.746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Baek J, Cho SY, Kang H, Ahn H, Jung WB, Cho Y et al. (2018) Distinct mechanosensing of human neural stem cells on extremely limited anisotropic cellular contact. ACS Appl. Mater. Interfaces 10, 33891–33900 10.1021/acsami.8b10171 [DOI] [PubMed] [Google Scholar]
  • 47.Gu H, Yu SP, Gutekunst CA, Gross RE and Wei L (2013) Inhibition of the Rho signaling pathway improves neurite outgrowth and neuronal differentiation of mouse neural stem cells. Int. J. Physiol. Pathophysiol. Pharmacol 5, 11–20 PMID:23525456 [PMC free article] [PubMed] [Google Scholar]
  • 48.Su YT,Lau SF, Ip JPK , Cheung K, Cheung THT, Fu AKY et al. (2019) alpha2-Chimaerin is essential for neural stem cell homeostasis in mouse adult neurogenesis. Proc. Natl Acad. Sci. U.S.A 116, 13651–13660 10.1073/pnas.1903891116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lian G, Wong T, Lu J, Hu J, Zhang J and Sheen V (2019) Cytoskeletal associated filamin A and RhoA affect neural progenitor specification during mitosis. Cereb. Cortex 29, 1280–1290 10.1093/cercor/bhy033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yang J, Wu C, Stefanescu I, Jakobsson L, Chervoneva I and Horowitz A (2016) Rhoa inhibits neural differentiation in murine stem cells through multiple mechanisms. Sci. Signal 9, ra76 10.1126/scisignal.aaf0791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kamishibahara Y, Kawaguchi H and Shimizu N (2016) Rho kinase inhibitor Y-27632 promotes neuronal differentiation in mouse embryonic stem cells via phosphatidylinositol 3-kinase. Neurosci. Lett 615, 44–49 10.1016/j.neulet.2016.01.022 [DOI] [PubMed] [Google Scholar]
  • 52.Katayama K, Melendez J, Baumann JM, Leslie JR, Chauhan BK, Nemkul N et al. (2011) Loss of rhoA in neural progenitor cells causes the disruption of adherens junctions and hyperproliferation. Proc. Natl Acad. Sci. U.S.A 108, 7607–7612 10.1073/pnas.1101347108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Cappello S, Attardo A, Wu X, Iwasato T, Itohara S, Wilsch-Brauninger M et al. (2006) The Rho-GTPase cdc42 regulates neural progenitor fate at the apical surface. Nat. Neurosci 9, 1099–1107 10.1038/nn1744 [DOI] [PubMed] [Google Scholar]
  • 54.Chen L, Liao G, Yang L, Campbell K, Nakafuku M, Kuan CY et al. (2006) Cdc42 deficiency causes Sonic hedgehog-independent holoprosencephaly. Proc. Natl Acad. Sci. U.S.A 103, 16520–5 10.1073/pnas.0603533103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Gehart H and Clevers H (2019) Tales from the crypt: new insights into intestinal stem cells. Nat. Rev. Gastroenterol. Hepatol 16, 19–34 10.1038/s41575-018-0081-y [DOI] [PubMed] [Google Scholar]
  • 56.Melendez J, Liu M, Sampson L, Akunuru S, Han X, Vallance J et al. (2013) Cdc42 coordinates proliferation, polarity, migration, and differentiation of small intestinal epithelial cells in mice. Gastroenterology 145, 808–819 10.1053/j.gastro.2013.06.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sakamori R, Das S, Yu S, Feng S, Stypulkowski E, Guan Y et al. (2012) Cdc42 and Rab8a are critical for intestinal stem cell division, survival, and differentiation in mice. J. Clin. Invest 122, 1052–1065 10.1172/JCI60282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Nalapareddy K, Hassan A, Sampson LL, Zheng Y and Geiger H (2021) Suppression of elevated Cdc42 activity promotes the regenerative potential of aged intestinal stem cells. Science 24, 102362 10.1016/jJsci.2021.102362 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Florian MC, Leins H, Gobs M, Han Y, Marka G, Soller K et al. (2020) Inhibition of Cdc42 activity extends lifespan and decreases circulating inflammatory cytokines in aged female C57BL/6 mice. Aging Cell 19, e13208 10.1111/acel.13208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Liu M, Zhang Z, Sampson L, Zhou X, Nalapareddy K, Feng Y et al. (2017) RHOA GTPase controls YAP-mediated EREG signaling in small intestinal stem cell maintenance. Stem Cell Rep. 9, 1961–1975 10.1016/j.stemcr.2017.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Myant KB, Scopelliti A, Haque S, Vidal M, Sansom OJ and Cordero JB (2013) Rac1 drives intestinal stem cell proliferation and regeneration. Cell Cycle 12, 2973–2977 10.4161/cc.26031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Myant KB, Cammareri P, McGhee EJ, Ridgway RA, Huels DJ, Cordero JB et al. (2013) ROS production and NF-kappaB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell 12, 761–773 10.1016/j.stem.2013.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Carmon KS, Gong X, Yi J, Wu L, Thomas A, Moore CM et al. (2017) LGR5 receptor promotes cell-cell adhesion in stem cells and colon cancer cells via the IQGAP1-Rac1 pathway. J. Biol. Chem 292, 14989–15001 10.1074/jbc.M117.786798 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zhang M, Wang X, Guo F, Jia Q, Liu N, Chen Y et al. (2019) Cdc42 deficiency leads To epidermal barrier dysfunction by regulating intercellular junctions and keratinization of epidermal cells during mouse skin development. Theranostics 9, 5065–5084 10.7150/thno.34014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zhan R, He W, Wang F, Yao Z, Tan J Xu R et al. (2016) Nitric oxide promotes epidermal stem cell migration via cGMP-Rho GTPase signalling. Sci. Rep 6, 30687 10.1038/srep30687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chai L, Cao C, Bi S, Dai X, Gan L, Guo R et al. (2010) Small Rho GTPase Rac1 determines human epidermal stem cell fate in vitro. Int. J. Mol. Med 25, 723–727 10.3892/ijmm_00000397 [DOI] [PubMed] [Google Scholar]
  • 67.Benitah SA and Watt FM (2007) Epidermal deletion of Rac1 causes stem cell depletion, irrespective of whether deletion occurs during embryogenesis or adulthood. J. Invest. Dermatol 127, 1555–1557 10.1038/sj.jid.5700738 [DOI] [PubMed] [Google Scholar]
  • 68.Benitah SA, Frye M, Glogauer M and Watt FM (2005) Stem cell depletion through epidermal deletion of Rac1. Science 309, 933–935 10.1126/science.1113579 [DOI] [PubMed] [Google Scholar]
  • 69.Yang S, Xin C, Zhang B, Zhang H and Hao Y (2020) Synergistic effects of Rho kinase inhibitor Y-27632 and noggin overexpression on the proliferation and neuron-like cell differentiation of stem cells derived from human exfoliated deciduous teeth. IUBMB Life 72, 665–676 10.1002/iub.2208 [DOI] [PubMed] [Google Scholar]
  • 70.Huang X, Chen X, Chen H, Xu D, Lin C and Peng B (2018) Rho/Rho-associated protein kinase signaling pathway-mediated downregulation of runt-related transcription factor 2 expression promotes the differentiation of dental pulp stem cells into odontoblasts. Exp. Ther. Med 15, 4457–4464 10.3892/etm.2018.5982 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hu JK, Du W, Shelton SJ, Oldham MC, DiPersio CM and Klein OD (2017) An FAK-YAP-mTOR signaling axis regulates stem cell-Based tissue renewal in mice. Cell Stem Cell 21, 91–106 e6 10.1016/j.stem.2017.03.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kan L, Smith A, Chen M, Ledford BT, Fan H, Liu Z et al. (2015) Rho-associated kinase inhibitor (Y-27632) attenuates doxorubicin-Induced apoptosis of human cardiac stem cells. PLoS ONE 10, e0144513 10.1371/journal.pone.0144513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Li C, Zhen G, Chai Y, Xie L, Crane JL, Farber E et al. (2016) Rhoa determines lineage fate of mesenchymal stem cells by modulating CTGF-VEGF complex in extracellular matrix. Nat. Commun 7, 11455 10.1038/ncomms11455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Arnsdorf EJ, Tummala P, Kwon RY and Jacobs CR (2009) Mechanically induced osteogenic differentiation-the role of RhoA, ROCKII and cytoskeletal dynamics. J. Cell Sci 122, 546–553 10.1242/jcs.036293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Thompson WR, Yen SS, Uzer G, Xie Z, Sen B, Styner M et al. (2018) LARG GEF and ARHGAP18 orchestrate RhoA activity to control mesenchymal stem cell lineage. Bone 107, 172–180 10.1016/j.bone.2017.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bryan BA, Li D, Wu X and Liu M (2005) The Rho family of small GTPases: crucial regulators of skeletal myogenesis. Cell. Mol. Life Sci 62, 1547–1555 10.1007/s00018-005-5029-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Qu P, Wang L, Min Y, McKennett L, Keller JR and Lin PC (2016) Vav1 regulates mesenchymal stem cell differentiation decision between adipocyte and chondrocyte via Sirt1. Stem Cells 34, 1934–1946 10.1002/stem.2365 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Liu X, Zhang Z, Yan X, Liu H, Zhang L, Yao A et al. (2014) The Rho kinase inhibitor Y-27632 facilitates the differentiation of bone marrow mesenchymal stem cells. J. Mol. Histol 45, 707–714 10.1007/s10735-014-9594-z [DOI] [PubMed] [Google Scholar]
  • 79.Umbayev B, Masoud AR, Tsoy A, Alimbetov D, Olzhayev F, Shramko A et al. (2018) Elevated levels of the small GTPase Cdc42 induces senescence in male rat mesenchymal stem cells. Biogerontology 19, 287–301 10.1007/s10522-018-9757-5 [DOI] [PubMed] [Google Scholar]
  • 80.Pattabiraman DR and Weinberg RA (2014) Tackling the cancer stem cells - what challenges do they pose? Nat. Rev. Drug Discov 13, 497–512 10.1038/nrd4253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Saygin C, Matei D, Majeti R, Reizes O and Lathia JD (2019) Targeting cancer stemness in the clinic: from hype to hope. Cell Stem Cell 24, 25–40 10.1016/j.stem.2018.11.017 [DOI] [PubMed] [Google Scholar]
  • 82.Prager BC, Xie Q, Bao S and Rich JN (2019) Cancer stem cells: the architects of the tumor ecosystem. Cell Stem Cell 24, 41–53 10.1016/j.stem.2018.12.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Clarke MF (2019) Clinical and therapeutic implications of cancer stem cells. N. Engl. J. Med 380, 2237–2245 10.1056/NEJMra1804280 [DOI] [PubMed] [Google Scholar]
  • 84.Rosenthal DT, Zhang J, Bao L, Zhu L, Wu Z, Toy K et al. (2012) Rhoc impacts the metastatic potential and abundance of breast cancer stem cells. PLoS ONE 7, e40979 10.1371/iournal.pone.0040979 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zheng L, Xiang C, Li X, Guo Q, Gao L, Ni H et al. (2018) 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 11, 72 10.1186/s13045-018-0613-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zanconato F, Cordenonsi M and Piccolo S (2016) YAP/TAZ at the roots of cancer. Cancer Cell 29, 783–803 10.1016/j.ccell.2016.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Elaimy AL, Guru S, Chang C, Ou J, Amante JJ, Zhu LJ et al. (2018) VEGF-neuropilin-2 signaling promotes stem-like traits in breast cancer cells by TAZ-mediated repression of the Rac GAP beta2-chimaerin. Sci. Signal 11, eaao6897 10.1126/scisignal.aao6897 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Fujimoto D, Ueda Y, Hirono Y, Goi T and Yamaguchi A (2015) PAR1 participates in the ability of multidrug resistance and tumorigenesis by controlling hippo-YAP pathway. Oncotarget 6, 34788–34799 10.18632/oncotarget.5858 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.He J, Bao Q, Zhang Y, Liu M, Lv H, Liu Y et al. (2018) Yes-associated protein promotes angiogenesis via signal transducer and activator of transcription 3 in endothelial cells. Circ. Res 122, 591–605 10.1161/CIRCRESAHA.117.311950 [DOI] [PubMed] [Google Scholar]
  • 90.Srinivasan S, Ashok V, Mohanty S, Das A, Das S, Kumar S et al. (2017) Blockade of Rho-associated protein kinase (ROCK) inhibits the contractility and invasion potential of cancer stem like cells. Oncotarget 8, 21418–21428 10.18632/oncotarget.15248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Liu C, Liu R, Zhang D, Deng Q, Liu B, Chao HP et al. (2017) MicroRNA-141 suppresses prostate cancer stem cells and metastasis by targeting a cohort of pro-metastasis genes. Nat. Commun 8, 14270 10.1038/ncomms14270 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Alkasalias T, Alexeyenko A, Hennig K, Danielsson F, Lebbink RJ, Fielden M et al. (2017) Rhoa knockout fibroblasts lose tumor-inhibitory capacity in vitro and promote tumor growth in vivo. Proc. Natl Acad. Sci. U.S.A 114, E1413–E1E21 10.1073/pnas.1621161114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Takeda H, Okada M, Suzuki S, Kuramoto K, Sakaki H, Watarai H et al. (2016) Rho-associated protein kinase (ROCK) inhibitors inhibit survivin expression and sensitize pancreatic cancer stem cells to gemcitabine. Anticancer Res. 36, 6311–6318 10.21873/anticanres.11227 [DOI] [PubMed] [Google Scholar]
  • 94.Pranatharthi A, Ross C and Srivastava S (2016) Cancer stem cells and radioresistance: Rho/ROCK pathway plea attention. Stem Cells Int. 2016, 5785786 10.1155/2016/5785786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wang JY, Yu P, Chen S, Xing H, Chen Y, Wang M et al. (2013) Activation of Rac1 GTPase promotes leukemia cell chemotherapy resistance, quiescence and niche interaction. Mol. Oncol 7, 907–916 10.1016/j.molonc.2013.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Yoon CH, Hyun KH, Kim RK, Lee H, Lim EJ, Chung HY et al. (2011) The small GTPase Rac1 is involved in the maintenance of stemness and malignancies in glioma stem-like cells. FEBS Lett. 585, 2331–2338 10.1016/j.febslet.2011.05.070 [DOI] [PubMed] [Google Scholar]
  • 97.Yoon C, Cho SJ, Chang KK, Park DJ, Ryeom SW and Yoon SS (2017) Role of Rac1 pathway in epithelial-to-mesenchymal transition and cancer stem-like cell phenotypes in gastric adenocarcinoma. Mol. Cancer Res 15, 1106–1116 10.1158/1541-7786.MCR-17-0053 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 98.Akunuru S, Palumbo J, Zhai QJ and Zheng Y (2011) Rac1 targeting suppresses human non-small cell lung adenocarcinoma cancer stem cell activity. PLoS ONE 6, e16951 10.1371/iournal.pone.0016951 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mizukawa B, O’Brien E, Moreira DC, Wunderlich M, Hochstetler CL, Duan X et al. (2017) The cell polarity determinant CDC42 controls division symmetry to block leukemia cell differentiation. Blood 130, 1336–1346 10.1182/blood-2016-12-758458 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Mizukawa B, Wei J, Shrestha M, Wunderlich M, Chou FS, Griesinger A et al. (2011) Inhibition of Rac GTPase signaling and downstream prosurvival Bcl-2 proteins as combination targeted therapy in MLL-AF9 leukemia. Blood 118, 5235–5245 10.1182/blood-2011-04-351817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Maldonado MDM and Dharmawardhane S (2018) Targeting Rac and Cdc42 GTPases in cancer. Cancer Res. 78, 3101–3111 10.1158/0008-5472.CAN-18-0619 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Shang X, Marchioni F, Evelyn CR, Sipes N, Zhou X, Seibel W et al. (2013) Small-molecule inhibitors targeting G-protein-coupled Rho guanine nucleotide exchange factors. Proc. Natl Acad. Sci. U.S.A 110, 3155–3160 10.1073/pnas.1212324110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kim JH, Park S, Lim SM, Eom HJ, Balch C, Lee J et al. (2020) Rational design of small molecule RHOA inhibitors for gastric cancer. Pharmacogenomics J. 20, 601–612 10.1038/s41397-020-0153-6 [DOI] [PubMed] [Google Scholar]
  • 104.Liu M, Bi F, Zhou X and Zheng Y (2012) Rho GTPase regulation by miRNAs and covalent modifications. Trends Cell Biol. 22, 365–373 10.1016/j.tcb.2012.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES