Next Article in Journal
Modelling of Neuronal Ceroid Lipofuscinosis Type 2 in Dictyostelium discoideum Suggests That Cytopathological Outcomes Result from Altered TOR Signalling
Next Article in Special Issue
The Roles of Hippo Signaling Transducers Yap and Taz in Chromatin Remodeling
Previous Article in Journal
Molecular Mechanisms Responsible for Therapeutic Potential of Mesenchymal Stem Cell-Derived Secretome
Previous Article in Special Issue
GPCR-Hippo Signaling in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of the Hippo Pathway in Fibrosis and Cancer

1
Department of Biomedical Sciences, Cancer Biology Graduate Program, Ajou University Graduate School of Medicine, Suwon 16499, Korea
2
Genomic Instability Research Center (GIRC), Ajou University School of Medicine, Suwon 16499, Korea
*
Author to whom correspondence should be addressed.
Submission received: 17 April 2019 / Revised: 10 May 2019 / Accepted: 14 May 2019 / Published: 16 May 2019
(This article belongs to the Special Issue Disease and the Hippo Pathway: Cellular and Molecular Mechanisms)

Abstract

:
The Hippo pathway is the key player in various signaling processes, including organ development and maintenance of tissue homeostasis. This pathway comprises a core kinases module and transcriptional activation module, representing a highly conserved mechanism from Drosophila to vertebrates. The central MST1/2-LATS1/2 kinase cascade in this pathway negatively regulates YAP/TAZ transcription co-activators in a phosphorylation-dependent manner. Nuclear YAP/TAZ bind to transcription factors to stimulate gene expression, contributing to the regenerative potential and regulation of cell growth and death. Recent studies have also highlighted the potential role of Hippo pathway dysfunctions in the pathology of several diseases. Here, we review the functional characteristics of the Hippo pathway in organ fibrosis and tumorigenesis, and discuss its potential as new therapeutic targets.

1. Introduction

The Hippo pathway is an evolutionarily conserved signaling transduction nexus that influences a wide range of biological processes during the growth and development of tissues and organs. This signaling pathway has recently been recognized as a master regulator of the malignant progression of many cancers by regulating cell proliferation and stem/progenitor-cell expansion [1]. Genetic studies in Drosophila provided the first identification of the Warts (Wts) as tumor suppressors [2,3].
Subsequent molecular studies further defined the details of core components of the Hippo pathway and their cellular functions, including cell growth, proliferation, survival, and organ-size control, demonstrating high levels of conservation from Drosophila to mammals. The Hippo pathway consists of a core kinases module and a transcriptional activation module. In Drosophila, the Hippo pathway includes a kinase cascade of Ste20-like kinase Hippo (Hpo) and NDR family kinase Wts [2,3,4,5,6,7,8]. Hpo forms a complex with the scaffolding protein Sav to phosphorylate and activate Wts, which physically associates with its regulatory protein Mob as Tumor Suppressor (Mats) [9,10,11]. Phosphorylated and activated Wts then inactivates Yorkie (Yki) via phosphorylation at serine residues [12], allowing it to interact with 14-3-3 and thereby accumulate in the cytosol [12,13,14,15]. The core components of the mammalian Hippo pathway include a kinase cascade of mammalian sterile 20-like kinase 1/2 (MST1/2) and large tumor suppressor kinase 1/2 (LATS1/2) [8,16,17,18]. MST1/2 and its regulatory protein SAV1 physically interact with and phosphorylate LATS1/2 kinase. Active LATS1/2 kinase in complex with Mob1 phosphorylates and inactivates the transcriptional co-activator Yes-associated protein (YAP) and transcriptional co-activator with a PDZ-binding motif (TAZ) to influence their subcellular localization in a 14-3-3-dependent manner [12,13,14,15,19]. YAP has also been identified as YES1-interacting protein, a tyrosine kinase, which is a key downstream effector of the Hippo pathway to function as a transcriptional co-activator [20]. TAZ (encoded by WWTR) is composed of a WW domain and PDZ-binding motif was demonstrated to play a key role as another mediator of the Hippo pathway similar to its paralog YAP [21]. Based on studies in Drosophila and mammals, the Hippo pathway kinase LATS1/2 regulates YAP/TAZ activity and their subcellular localization by direct phosphorylation of five consensus HX(R/H/K)XX(S/T) motifs [22,23,24,25]. Phosphorylated YAP/TAZ associate with 14-3-3 protein, leading to their cytoplasmic accumulation and the consequent loss of transcriptional co-activator function. Cytosolic YAP/TAZ are further phosphorylated by CK1 and LATS1/2, and are then rapidly destroyed by the ubiquitin-proteasome and lysosome system [26]. Conversely, unphosphorylated YAP/TAZ translocate to the nucleus and bind to the TEAD family transcription factors to activate the transcription of target genes that promote cell proliferation and inhibit apoptosis [27,28]. Besides TEADs, YAP/TAZ can interact with several other proteins, including Smad, Runx family, p73, ErbB4, Pax3, TTF1, PPARγ, and TBX5, which are responsible for transcriptional activation, and VGLL4, which is responsible for transcriptional repression [29].
Despite these functional similarities of YAP and TAZ, they nevertheless differ in their physiological and developmental functions. Much progress has been made in identifying the multiple new regulators of the Hippo pathway. Moreover, accumulating evidence, both physiological and pathological, points to a role of the central upstream Hippo kinase cascade in regulating the downstream effectors YAP/TAZ that sense extracellular environment signals, and integrate intracellular signaling to maintain a variety of biological functions, including tissue homeostasis and cancer [30,31,32]. Here, we review this research progress of the specific roles of YAP/TAZ to provide new insight into these important mediators of the Hippo pathway as effectors of fibrosis and cancer progression.

2. The Hippo Pathway in Organ Fibrosis

Myofibroblasts are responsible for fibrogenesis and produce a series of extracellular matrix (ECM) components such as collagens, laminins, and fibronectins to promote their contractile force. Recently, the Hippo pathway was shown to contribute to the pathogenesis of fibrosis, in which hyperactive YAP/TAZ accumulate in both the epithelial and stromal tissue compartments of fibrotic tissues.

2.1. Lung Fibrosis

The basic unit of the lungs is the pulmonary alveoli comprised of epithelial and ECM layers surrounded by capillaries [33]. During lung development and repair, respiratory epithelial and mesenchymal progenitors show dramatic changes in their cellular behavior to maintain the appropriate cell type composition and structural organization of the lung epithelium through a re-epithelialization process [34]. The alveolar epithelial cells are the main source of fibroblasts and myofibroblasts, which convert to a mesenchymal cell phenotype following injury, and could trigger the development of fibrosis through a process known as epithelial-mesenchymal transition (EMT). The Hippo pathway controls epithelial progenitor cell proliferation, migration, and differentiation in the developing and mature lungs [35,36].
TAZ is proposed to act as a co-activator of various transcriptional factors through binding of the WW domain to the (L/P)PXY motif of transcriptional factors. During fetal lung development, TAZ and thyroid transcription factor 1 (TTF-1) are co-expressed in respiratory epithelial cells, and TAZ directly interacts with the (L/P)PXY motif of TTF-1 to activate the transcription of target genes, including surfactant protein C (SP-C) that maintains lung morphogenesis [37]. Moreover, Mitani et al. [38] showed that TAZ knockout mice exhibited abnormal lung alveolarization. Microarray analysis of wild-type and TAZ-deficient mouse lungs revealed that connective tissue growth factor (Ctgf), a known direct TAZ-TEAD target gene, is the main player in peripheral epithelial cell differentiation and lung development. Previous studies have also suggested that CTGF promotes EMT, lung fibrosis, and lung development, implying a correlation between TAZ and CTGF in lung physiological and pathological functions [39,40,41].
Moreover, the matrix mechanical environment influences the subcellular localization of YAP/TAZ in lung fibroblasts [42]. An active YAP/TAZ positive feed-forward mechanism was shown to amplify a profibrotic response to induce fibrosis in the lung tissue of mice following tail vein injection of YAP 5SA/TAZ 4SA-overexpressing fibroblasts. In a similar fashion, YAP and TAZ were found to be highly expressed in the fibroblastic foci of the lungs from patients with idiopathic pulmonary fibrosis (IPF) with obvious nuclear expression of YAP/TAZ, indicating that YAP/TAZ play a significant role in fibrosis [43]. When cultured on a stiff matrix, YAP/TAZ accumulated in the nucleus, and the lung fibroblasts exhibited increased proliferation, contraction, and ECM production, comparable to the results for cells cultured on a soft matrix. However, silencing of YAP and TAZ by small interfering RNAs (siRNAs) reduced the fibroblast responses induced by a stiff matrix, suggesting that YAP and TAZ activation are indispensable for these phenotypic features of myofibroblasts. In further support of the association of the function of YAP/TAZ with lung fibrosis, Liu et al. showed that the forced expression of a constitutively active form of YAP and TAZ in lung fibroblasts increased the expression of CTGF and PAI-1, also known as SERPINE1, along with that of ECM-related proteins such as collagens and fibronectin on a stiff matrix. However, an active TAZ mutant could not induce the expression of these ECM proteins in cells cultured on a soft matrix. These findings suggested that both mechanical stress and fully activated TAZ are necessary to induce the expression of profibrotic genes in the development of tissue fibrosis [44]. RNA sequencing identified that the genes that are positively regulated by TAZ in lung fibroblasts are linked to cell migration and motility, and are closely related to genes regulated by transforming growth factor (TGF)-β, a central mediator in lung fibrosis [43]. In addition, the profibrotic effect of YAP/TAZ was demonstrated in murine models of pulmonary fibrosis. Deletion of a single copy of Taz (WWTR1) attenuated bleomycin-induced pulmonary fibrosis and diminished collagen deposition and lung elastance, indicating that TAZ is a key molecule in fibrogenic events [38]. More recently, emerging evidence suggests the involvement of microRNA (miRNA) in IPF progression. For example, YAP1/Twist-induced fibroblast activation and fibrogenesis were blocked by miR15a [45]. IPF is a progressive chronic interstitial lung disease and is usually a fatal condition with no available cure [46]. IPF is characterized by fibroblast proliferation, remodeling of the ECM, and the contraction of fibroblasts, which contribute to tissue tension or stiffness that results in irreversible distortion of the lung architecture [47]. Single-cell RNA profiles from both normal and IPF lung epithelial cells demonstrated that gene expression patterns of IPF cells were in a multilineage-like state, which was not detected during normal lung development [48]. Transcriptomic analysis of IPF cells identified abnormal activation of several pathways that regulate the multilineage differentiation program, including TGF-β, Hippo, PI3K/AKT, p53, and Wnt signaling cascades. Nuclear YAP localization was shown to be increased in IPF lung epithelial cells [49]. YAP cooperates with mTOR-PI3K-AKT signaling to activate aberrant cell proliferation and migration, and inhibits the epithelial cell differentiation that may be involved in the development of IPF. Thus, mechanistically, YAP/TAZ activity contributes to the pathogenesis of IPF in both lung fibroblasts and epithelial cells. Indeed, Speight et al. showed that alpha-smooth muscle actin (α-SMA) expression is regulated by TAZ in the injured epithelium during phenotypic changes such as the epithelial–myofibroblast transition [50]. Similarly, recent reports demonstrated that stiff matrices activated fibroblasts and hepatic stellate cells (HSCs) to promote matrix synthesis through YAP/TAZ-mediated gene induction, including CTGF and PAI-1 [35,43]. Taken together, these observations suggest that YAP and TAZ are important mechanosensors of TGF-β-dependent and -independent lung fibrogenesis (Figure 1a).

2.2. Kidney Fibrosis

The kidney comprises numerous distinct cell types that are organized into a renal filter, including glomerular endothelial cells, glomerular basement membrane, and podocytes [51]. In mice, YAP/TAZ are essential for epithelial cell growth and establishing the proper kidney architecture [52,53,54]. Several studies have demonstrated the involvement of YAP/TAZ in the progression of cystic kidney disease (CKD) in patients and in a mouse model based on their enhanced nuclear localization in epithelial cells [55,56,57]. Various renal injuries resulting from IgA nephropathy or membranous nephropathy lead to renal tubulointerstitial fibrosis (TIF), which is the end stage of kidney failure known as progressive fibrotic chronic kidney disease. TAZ protein levels were shown to be markedly increased in both mouse Sav1-depleted kidneys after unilateral ureteral obstruction (UUO) and in patients with TIF/CKD [58]. High-level expression and nuclear accumulation of TAZ in the renal tubulointerstitium were demonstrated in three mouse models of renal injury [59]. Consistent with these findings, the YAP protein levels increased in both the cytoplasm and nucleus in regenerative and poorly differentiated renal tubules during the transition from acute kidney injury to CKD [60]. In addition, activation of YAP/TAZ after UUO led to TGF-β-induced EMT-like features in renal TIF [58]. As expected, deletion of Sav1 in renal tubular epithelial cells induced YAP/TAZ activation and increased the EMT phenotype via induction of SNAI2 and a-SMA, which may improve renal fibrosis.
Other studies have demonstrated that the Hippo pathway plays a crucial role in tissue repair after renal injury and is involved in maintaining the kidney filtration barrier (Figure 1b) [53,61,62]. Podocyte-specific deletion of Yap1 in mice induced cell death and the progression of significant proteinuria and glomerular lesions [63]. In kidney fibrosis, TGF-β-induced profibrotic Smad signaling by controlling Smad2/3 localization is regulated by stiffening of the ECM, and this mechanosensing is mediated by YAP/TAZ in renal fibroblasts [64]. The abundance and activation of YAP/TAZ were detected in podocytes of a puromycin aminonucleoside-induced injury model of glomerular disease [62]. Gene expression analysis in the podocytes further indicated that overexpressed YAP upregulated ECM constituents (i.e., COL6A1, BCAM, and ADAMTS1) and growth factor-related proteins (i.e., PLOD2, PDAP1, and PRMT5), which changed the mechanical properties of the glomerular basement membrane, a prognostic marker of glomerular disease progression.

2.3. Liver Fibrosis

The Hippo pathway also plays an important role in liver development [65]. Yap overexpression or Mst1/2-knockout in the liver and other upstream regulators causing hyperactivation of YAP was shown to contribute to hepatomegaly and liver tumorigenesis [14,66,67,68]. Large livers comprise activated progenitor cells, hyperproliferating hepatocytes, and bile duct cells, and are more likely to develop liver cancer at later stages [14,66,68]. Thus, the Hippo signaling pathway may control the activation of progenitor cells and their differentiation towards the hepatocyte lineage. HSCs are the predominant mediators of liver fibrosis/cirrhosis and are rapidly activated and transdifferentiate into fibrogenic hepatic myofibroblasts during liver injury. HSCs from a CCl4-induced mouse liver fibrosis model and from the livers of patients with hepatitis C virus infection showed predominant nuclear YAP localization, which could be used for staging fibrosis [69]. High abundance of nuclear YAP denotes the activation of YAP in myofibroblasts. YAP inhibition by specific siRNA or treatment with verteporfin (VP), which disrupts the YAP/TAZ-TEAD interaction, decreased target gene expression and prevented the transdifferentiation of quiescent HSCs into myofibroblasts. YAP has been confirmed as a critical driver of HSC activation based on studies with in vivo and in vitro models of liver fibrosis. Consistent with this concept, Hedgehog signaling-mediated YAP activation was found to be necessary for HSC transdifferentiation and proliferation by a glutaminolytic process [70]. TAZ expression is elevated in hepatocytes of the liver of patients with non-alcoholic steatohepatitis (NASH) and in NASH model mice [71].
The key mechanism driving the transition from steatosis to NASH is TAZ-mediated Indian hedgehog (Ihh) gene induction, which enhances transcriptional reprogramming by modulating profibrotic gene expression in HSCs, the fibrogenic precursor cells of ECM-producing myofibroblasts. In the same study, treatment of TAZ-specific siRNA to the hepatocytes isolated from NASH mouse model suppressed hepatic inflammation, fibrosis, and cell death, whereas reconstituted TAZ expression promoted the progression from steatosis to NASH (Figure 1c).

2.4. Heart Fibrosis

The adult mammalian heart has restricted regenerative capacity and repair potential. Thus, after injury or disease, a large number of cardiomyocytes are lost and a fibrotic scar is formed, which can result in heart failure [72]. Conversely, the neonatal heart has potential for enhanced cardiomyocyte proliferation. Therefore, elucidation of the regulatory mechanisms underlying cardiomyocyte proliferation may provide new insight for regenerative repair strategies. Several studies have also demonstrated a role of the Hippo pathway in regulating cardiac development. During mouse embryogenesis, loss of Mst1/2 or Lats, parts of the central kinase cascade, induced cardiomyocyte proliferation [73]. Similarly, in damaged hearts, inactivated Salv and Lats led to re-entry of the mitotic cell cycle and subsequent cytokinesis, indicating that Hippo signaling is a major regulator of cardiomyocyte renewal and regeneration [74]. A constitutively active S112A mutant form of YAP enhanced cardiomyocyte proliferation and heart growth, whereas cardiac-specific deletion of Yap decreased cardiomyocyte proliferation in the embryonic and postnatal mouse heart [75,76]. Consistent with these findings, deletion of Yap and Taz was shown to result in lethal cardiomyopathy, whereas activated Yap stimulated cardiac regeneration and enhanced contractility after myocardial infarction in the adult mouse heart [77]. Insulin-like growth factor signaling and the Wnt pathway cooperatively contribute to the beneficial effects of YAP on fetal cardiac growth, homeostasis, and normal function of the postnatal heart [73,75,78]. In addition, Angiotensin II (ANG II) induced cardiovascular fibrosis in a YAP/TAZ-dependent manner. Lovastatin mediated YAP/TAZ suppression decreased ANG II-induced fibrogenic gene expression as well as cardiovascular fibrosis [79].

2.5. Skin Fibrosis

YAP also plays an important role in maintaining normal skin homeostasis in response to extracellular cues by promoting cell proliferation in epidermal stem and progenitor cells, and tissue expansion [80,81] Genetic analysis showed that YAP is highly expressed and shows nuclear localization in epidermal progenitor and stem cells, revealing a requirement for TEAD to mediate YAP’s functions in controlling their self-renewal and maintaining their undifferentiated state [82]. Moreover, enhanced nuclear YAP/TAZ levels have been detected in the dermis during the skin wound-healing process. Downregulation of YAP/TAZ expression delayed healing and reduced the expression levels of TGF-β and its target genes such as p21 and Smad family members [83]. The expression and activation of YAZ/TAZ are increased in Dupuytren disease tissues, which are characterized by the formation of myofibroblast-rich cords and nodules in the hands [84]. This study further showed that YAP1 is involved in the differentiation of human dermal fibroblasts to myofibroblasts, and is necessary for maintenance of a smooth muscle cell-like contractile phenotype in primary Dupuytren myofibroblasts. In addition, YAP/TAZ levels were found to be elevated in systemic sclerosis (SSc) biopsy sections, mainly localized within the nucleus, and they mediated the profibrotic responses in dermal fibroblasts [85]. In mice, depletion of YAP/TAZ was effective in inhibiting bleomycin-induced fibrosis, a model of SSc dermal fibrosis, via inhibition of the PI3K-AKT-GSK3-β pathway. Mechanistically, dimethyl fumarate significantly blocked nuclear YAP localization in SSc dermal fibroblasts and prevented bleomycin-induced skin fibrosis (Figure 1d).

2.6. Tumor Fibrosis

In normal conditions, myofibroblasts modulate the wound-healing process and participate in tissue remodeling via producing a large amount of ECM, matrix metalloproteinases (MMPs), and tissue inhibitors of metalloproteinases (TIMPs) [86]. Myofibroblast activation ultimately occurs via MMPs and TIMPs-mediated ECM reorganization and contraction, conferring the contractile properties of myofibroblasts containing contractile actin filament bundles. In solid tumors, cancer cells reciprocally interact with non-cancerous stromal cells and the ECM to create a permissive tumor microenvironment for proliferation and maintenance of cancer cell-like features [87,88,89,90]. In particular, fibroblasts that generally reside in the tumor microenvironment are denoted as cancer-associated fibroblasts (CAFs). During tumorigenesis, both cancer cells and infiltrated immune cells release various cytokines, growth factors, and mediators to stimulate the transdifferentiation and activation of CAFs [91]. CAFs were also suggested to facilitate cancer cell invasion and metastasis through the production of soluble factors and remodeling of the ECM. A co-culture model of cancer cells and stromal fibroblasts revealed that both protease and force-mediated ECM remodeling enabled the invasion of cancer cells via a mechanism requiring CAFs [92]. CAFs mainly promote cancer cell invasion by altering the organization and physical properties of the basement membrane, leading to the formation of gaps that allow for migration [93]. For instance, mRNA expression profiling analyses showed that YAP activation is critical for the establishment and maintenance of CAFs, and that its function is necessary for ECM stiffness modulation in the progression of mammary tumors [94]. YAP/TAZ upregulation is a representative marker of high-grade breast cancer, which is required for CAFs to induce ECM stiffening, angiogenesis, and invasion of cancer cells [95]. Remarkably, increased ECM stiffness promoted cellular YAP activity to induce its target genes, and therefore support their maintenance in the tumor microenvironment by regulating matrix stiffening. Furthermore, ECM stiffness and YAP/TAZ activity function via a reinforcing feed-forward loop to preserve the CAF phenotype. Strikingly, the MRTF-SRF pathway is activated in CAFs and is required for its contractility and pro-invasive properties [96]. Activation of the MRTF-SRF pathway, which is also mechano-responsive and mandatory for maintenance of the CAF phenotype, is dependent on the transcription co-activator YAP. MRTF and YAP can indirectly activate each other through their ability to modulate actin cytoskeletal dynamics and tumor invasion under control of Rho GTPase signaling.

2.7. The Links between Fibrosis and Cancer via the Hippo Pathway

The wound-healing process in injured tissue results in the infiltration of stromal cells, including immune cells, endothelial cells, pericytes, adipocytes, and fibroblasts, to the local tissue. This results in the release of cytokines and growth factor from infiltrated immune cells, which triggers the activation and transdifferentiation of fibroblasts into myofibroblasts. Upon injury, changes to the microenvironment activate myofibroblasts to secrete several growth factors, chemokines, and ECM components, including fibronectin, collagens, laminin, fibronectin, and tenascin [86,97]. In the case of a chronic injury that causes long-term inflammation, myofibroblasts rapidly increase the production of ECM, resulting in excessive deposition and resultant fibrosis. Stressed myoblasts are key actors in fibrosis development, and act as drivers in metastasis and invasion during tumor progression, thus representing the main link between fibrosis and cancer development [91,98,99,100]. Fibroblast-mediated ECM changes in the stroma may trigger a dramatic alteration in the Hippo pathway in both fibrosis and tumor progression (Figure 2). Recent findings imply that ECM cues and YAP/TAZ collaborate to operate a positive feedback circuit in cancer cells that contributes to matrix stiffening, resulting in further activation of YAP/TAZ oncogenic activity.

3. The Hippo Pathway in Cancer Progression

3.1. Genetic and Epigenetic Altherations in the Hippo Pathway Genes

Actively proliferating cells or cancer cells typically show a common feature of highly enriched YAP/TAZ protein in the nucleus, which exists in a dephosphorylated form and can enhance fundamental cellular functions, including cell proliferation and tumorigenesis [101]. Extensive research implicates dysregulation of the Hippo pathway in a wide spectrum of human cancers across the body [7,102]. However, only limited cancers have been associated with somatic mutations in components of the Hippo pathway compared with those of key growth and oncogenic signaling pathways. For instance, NF2, an upstream regulator of the Hippo pathway, is an extensively studied tumor suppressor, and a high frequency of NF2-inactivating mutations is a characteristic of several human cancers, including acoustic neuromas, meningiomas, schwannomas, and mesothelioma [103,104,105]. Interestingly, the core MST1/2-LATS1/2 kinase module functions as a tumor suppressor in transgenic and knockout mouse models, and no somatic mutation in these genes has been discovered in human cancer to date. However, methylation-mediated epigenetic mechanisms contribute to downregulation of LATS1/2, MST1/2, and its regulator RASSF1 in some cancer types [106,107,108]. This suggests that loss-of-function mutations in tumor suppressor genes may be mediated by non-mutational mechanisms [67,68,108,109,110,111,112]. Mutation of the YAP gene at the chromosome 11q22 amplicon has been reported in various human cancers [113,114]. Recently, a YAP and transcription factor E3 (TFE3) (YAP-TFE3) fusion gene, and a TAZ and calmodulin-binding transcription activator 1 (CAMTA1) (TAZ-CAMTA1) fusion gene were reported in cases of epithelioid hemangioendothelioma, a rare type of soft tissue sarcoma [115,116]. Although the effects of these fusions on the functions of the translated proteins in cancer development and progression remain to be determined, they may be functionally related to the transcriptional regulatory properties ascribed to both TAZ-CAMTA1 and YAP-TFE3.

3.2. YAP/TAZ as Mediators of the Mechanical Cues Shaping the Tumor Microenvironment

To respond to changes in the extracellular environment, cells harmonize an intracellular signaling network to achieve the desired cellular behavior during physiological growth and regeneration. ECM mechanical cues are powerful determinants of cell proliferation, differentiation, and cell death, acting as potent regulators of YAP/TAZ. For instance, the cell-cell contact resulting from a high cell density activates the growth inhibition signaling pathway that is predominantly mediated by the Hippo pathway [22,117,118]. As the cell density increases, LATS1/2 are phosphorylated and activated by cell-to-cell contact, leading to the inactivation of YAP/TAZ, which constitutes a central mechanism for contact inhibition. This contact-mediated inhibition in turn regulates YAP/TAZ-TEAD-mediated transcription, which is also important for embryo development [119]. Dupont et al. [120] first reported that the intracellular localization and activity of YAP/TAZ correlate with ECM rigidity and cell spreading. For example, YAP/TAZ translocate to the nucleus and function as transcriptional co-activators in response to exposure to a stiff ECM, and can stimulate the proliferation and survival of cancer cells. However, on soft matrices, cells are rounded and minimally adhesive by limiting the adhesive area. As expected, the main subcellular localization of YAP/TAZ is the cytoplasm. YAP/TAZ are downstream effectors of RhoA-ROCK-mediated actin cytoskeleton rearrangements, and thus regulate development and tumorigenic processes. In particular, a comprehensive study suggested that LATS1/2-dependent and -independent mechanisms synergize mechanical cues mediated by YAP/TAZ activation. When a cell detaches from the ECM, it undergoes the apoptotic process of anoikis owing to a dysregulated cytoskeleton and increased LATS1/2 kinase activity, which in turn inhibits YAP/TAZ activity [121].
Furthermore, Hippo pathway activity and YAP/TAZ localization depend on cytoskeletal dynamics in response to a mechanical input, including cell spreading, stretching, size, and shape [120,122,123]. Integrin-mediated cell adhesion to the ECM and association with intracellular actin filaments is required for many physiological functions [124]. Focal adhesions comprising focal adhesion kinase (FAK), Src family kinases, and integrins act as a sensor of the physical properties of the environment surrounding the ECM through forces exerted on the adhesions and via intracellular signals in response to extracellular physiochemical stimuli. FAK activates the RhoA-ROCK signaling pathway that controls remodeling of the actin cytoskeleton, which increases the stress fiber assembly, large microfilament bundles, and cell contractility (Figure 3c). In addition to FAK, ILK has been implicated in integrin-mediated signaling and promotes the inhibitory phosphorylation of MYPT1, leading to inactivation of Merlin, which is a key upstream regulator of the Hippo pathway (Figure 3a) [125]. This integrin-mediated FAK–Src signaling triggers YAP/TAZ activation. Moreover, integrin-dependent cell adhesion induces p21-activated kinase 1 (PAK1) activation, which is required for YAP/TAZ nuclear localization in a Merlin-dependent manner (Figure 3b). The small GTPase RAC1 and its downstream effector PAK1 promote the direct phosphorylation of NF2, which potentiates the physical interaction between NF2 and YAP, resulting in the nuclear translocation of activated YAP/TAZ [126]. In addition, fibronectin adhesion-induced stimulation of the FAK-Src-PI3K pathway acts as an upstream negative regulator of the Hippo pathway, which leads to activation of YAP/TAZ in a LATS1/2 kinase-dependent manner [127]. Interestingly, the surrounding ECM activates YAP/TAZ directly to promote the transcription of genes involved in cytoskeleton remodeling through the RhoA-ROCK pathway [128].
Morphological manipulation without cell–cell contact and changes in the number of stress fibers according to the adhesion area enhances the nuclear localization of YAP through activation of the Hippo pathway [129]. Moreover, deficiency of destrin, an actin-depolymerizing factor, leads to the induction of F-actin polymerization and the consequent hyperproliferation of corneal epithelial cells [130]. In contrast, reduction of the F-actin binding protein Cofilin, CapZ, and gesolin, which are essential elements of the actin cytoskeleton, aberrantly activates F-actin polymerization and therefore restricts activation of the Hippo pathway to in turn positively regulate YAP/TAZ activity [123]. Agrin, a proteoglycan, promotes liver carcinogenesis as an ECM sensor, leading to enhancement of YAP stability and activity through the combinatory activation of integrin-FAK- and Lrp4/MuSK receptor-mediated signaling pathways (Figure 3d). In this context, Agrin transduces stiffness signals via RhoA-dependent actin cytoskeletal rearrangements. Stimulated ILK-PAK1 signaling antagonizes Merlin and LATS1/2 activation [131,132,133]. Glypican-3, a member of the glypican family of heparan sulfate proteoglycans, is highly and specifically expressed in hepatocellular carcinoma (HCC) [134]. Glypican-3 targeting HN3 was shown to significantly inhibit the growth of HCC cells and xenograft tumors in nude mice by inhibiting YAP function. Disturbed fluid flow is widely recognized to induce the expression of YAP/TAZ target genes through the coordination of Rho GTPase activities, highlighting YAP/TAZ as a key node in mediating mechanical cues and maintaining vascular homeostasis [135,136,137]. Moreover, the mechanical stability of the nucleus dictates YAP nuclear entry [138], and a mechano-sensing mechanism has been identified to be directly mediated by nuclear pore complexes. On a stiff substrate, focal adhesions and stress fibers lead to nuclear flattening, which stretches the nuclear pores by transducing mechanical forces. These stretched and curved nuclear pores induce YAP import due to the relaxation of mechanical restriction to nuclear pore complexes.
Overall, Rho GTPase and cytoskeletal tension are responsible for the transduction of mechanical stress in cells and have substantial effects on YAP/TAZ activity. For example, an F-actin disruptor and Rho GTPase inhibitor considerably affect YAP nuclear translocation and activity [120,121,129]. However, it remains unclear precisely how the actin cytoskeleton and mechanical stress converge on YAP/TAZ regulation. Detachment of cells from the ECM triggers LATS1/2, lead to inactivation of YAP/TAZ in a cytoskeleton- or JNK-dependent manner [129,139]. Accordingly, the detailed mechanisms underlying involvement of the Hippo pathway kinases MST1/2 and LATS1/2 as mediators to connect mechanical cue remain to be fully elucidated. Beyond ECM remodeling, CAFs are also capable of secreting a wide range of mediators, including growth factors, chemokines, MMPs, TIMPs, and ECM components [100]. These secreted mediators can act as ligands for receptors expressed by other cell types in the tumor microenvironment, which initiate the signaling cascades that activate YAP/TAZ and lead to tumor progression. For example, in response to epidermal growth factor, its receptor regulates the Hippo pathway through the PI3K-PDK1 pathway, resulting in inactivation of LATS1/2 and activation of YAP [140]. Vascular endothelial growth factor, a major driver of blood vessel formation, activates YAP/TAZ via actin cytoskeleton dynamics to facilitate a transcriptional program to further control cytoskeleton dynamics and initiate a proper angiogenic response [141]. Fibroblast growth factor 5-driven fibroblast growth factor receptor (FGFR) signaling promotes YAP activation, setting off a feed-forward loop whereby YAP forms a complex with TBX5 and induces the expression of FGFR1, 2, and 4 in human cholangiocarcinoma cell lines (Figure 4d) [142]. YAP and KRAS coordinately regulate the cell cycle and DNA replication program by forming a complex with the transcription factor E2F in pancreatic ductal adenocarcinoma (Figure 4g) [143]. Jang et al. (Figure 4a) [144] showed that mechanical stress regulates the cell cycle via the Hippo pathway. In breast cancer cells, YAP and TEAD directly bind to the Skp2 promoter to enhance its transcription upon receiving a mechanical cue, which is necessary for cell cycle progression. Interaction of YAP with mutant p53 together form a complex with NF-Y, a nuclear transcription factor, which upregulates the expression of cell cycle genes, including cyclin A (CCNA), cyclin B (CCNB), and CDK1, in breast cancer cells (Figure 4b) [145]. YAP also enhances malignant mesothelioma cell growth and survival via the upregulation of cyclin D1 (CCND1) and FOXM1 in cooperation with TEAD (Figure 4a) [146]. Furthermore, YAP/TAZ/TEAD cooperate with AP-1 in the transcriptional amplification of the c-Myc oncogene to promote cell cycle progression in skin cancer (Figure 4c) [147].

3.3. YAP/TAZ Signaling in the EMT

The EMT is a complex transdifferentiation process that allows for epithelial cells to acquire the stem-like properties of mesenchymal cells, and is an important process in both fibrosis and cancer metastasis. The EMT is executed through a series of transcription factor networks, including TWIST1/2, ZEB1/2, Snail, and Slug. During cancer progression, cancer cells acquire aggressive features such as migration, invasion, chemoresistance, and apoptosis resistance that are all triggered by the EMT [148]. YAP/TAZ signaling has also been shown to play a role in the EMT in mammary epithelial cells. Constitutively active TAZ was shown to promote cell proliferation and EMT [25]. Moreover, YAP levels were found to be elevated in the stem cell-like cells derived from non-small lung cancer cells, and contributed to their self-renewal through Sox2. These effects required a physical interaction between YAP1 and Oct4 (Figure 4e) [149]. YAP and KRAS act cooperatively with the transcription factor FOS to activate Slug and vimentin-mediated EMT (Figure 4f) [150]. TAZ activity is required for the self-renewal and tumorigenesis of breast cancer cells. Twist and Snail-mediated EMT processes induce Scribble delocalization so that TAZ is released from Scribble, which stimulates the accumulation of stem-like progenitor cells in breast cancer [151]. These findings suggest that TAZ-induced EMT functions as a feed-forward mechanism of TAZ activation. The direct interaction between ZEB1 and YAP, but not TAZ, stimulated the transcription of a ZEB1/YAP target gene set that is known to promote cancer aggressiveness (Figure 4h) [152]. Indeed, this target gene set was found to be an excellent predictor of poor clinical outcomes in patients with particularly hormone receptor-negative cancer. Similarly, Snail/Slug-YAP/TAZ complexes control YAP/TAZ protein levels and the expression of target genes that modulate self-renewal, differentiation, and bone formation (Figure 4i) [153]. Therefore, crosstalk between YAP/TAZ and EMT transcription factors such as ZEB1/2, Snail/Slug, and Twist modulates the EMT programs, thereby acquiring the characteristics of malignant cancer cells. A genome-wide expression study revealed that TAZ protein levels were higher in breast cancer stem cells compared with those of differentiated breast cancer cells. A high level of TAZ was also associated with the chemoresistance and migratory potential of breast cancer stem cells [154]. The YAP-TEAD complex can directly bind to the SOX promoter region to upregulate SOX9 levels and induced a cancer stem cell phenotype in esophageal cancer cells (Figure 4a) [155]. YAP and the COX2 signaling pathway coordinately regulate urothelial cancer cells with stem cell-like properties via SOX2 induction [156]. Conversely, SOX2 maintains cancer stem cells in osteoblastoma by direct inhibition of NF2 and WWC1, upstream activators of the Hippo pathway, leading to activation of YAP [157] (Figure 4).

4. Conclusions

Accumulating evidence has highlighted the pivotal role of the Hippo pathway and its contribution to organ and tissue homeostasis under various chemical and physical signals. The MST1/2-LATS1/2 kinase cascade incorporates various upstream inputs to regulate YAP/TAZ activity and subcellular localization so as to maintain balanced processes, including physiological growth, development, and regeneration. In recent years, increasing research efforts have focused on unmasking the physiological/pathological functions of the Hippo pathway using animal models and patients, revealing a critical role in both fibrosis and cancer, which are inextricably linked. YAP/TAZ acts as a cellular mechanosensor that induces the transdifferentiation of fibroblasts to myofibroblasts in tissue fibrosis. In response to matrix stiffness, YAP/TAZ activation drives profibrotic and tumorigenic effects. YAP/TAZ signaling is also capable of initiating the EMT via EMT-related transcription factors, and this YAP/TAZ-dependent EMT program during tumorigenesis may further stimulate stemness and tumorigenicity.
These new insights into the diverse functions of the Hippo pathway will help in discovering novel therapeutic targets for the treatment of fibrosis and cancer. For example, VP is a small molecule that modulates the YAP and TEAD interaction and could reverse fibrosis. [49,64,69]. Given the clear profibrotic effect of YAP/TAZ, such pharmacological inhibitors targeting the Hippo pathway may be beneficial for treating both fibrosis and cancer (Table 1) [69,79,158,159,160,161,162,163,164,165]. However, targeting the Hippo pathway may also cause undesired harmful effects because of its central roles in organ development, homeostasis, and regeneration. Therefore, further studies to uncover the other mechanisms or unknown molecules regulating YAP/TAZ activity are expected to offer promising therapeutic targets in fibrosis and cancer progression.

Author Contributions

Conceptualization, J.M. and C.K.; Writing—Original Draft Preparation, J.M., C.K. and S.C.; Writing—Review & Editing, J.M. and C.K.; Visualization, S.C.; Funding Acquisition, J.M.

Acknowledgments

We apologize for many of most important research contributions to the Hippo pathway, fibrosis, and cancer fields that we could not cite in this review. This work is supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, Ict and Future Planning [NRF-2016R1C1B2016135 (Young Researcher Program)].

Conflicts of Interest

The authors declare that there is no conflict of interest.

References

  1. Yu, F.X.; Zhao, B.; Guan, K.L. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell 2015, 163, 811–828. [Google Scholar] [CrossRef] [PubMed]
  2. Harvey, K.F.; Pfleger, C.M.; Hariharan, I.K. The Drosophila Mst ortholog, hippo, restricts growth and cell proliferation and promotes apoptosis. Cell 2003, 114, 457–467. [Google Scholar] [CrossRef]
  3. Jia, J.; Zhang, W.; Wang, B.; Trinko, R.; Jiang, J. The Drosophila Ste20 family kinase dMST functions as a tumor suppressor by restricting cell proliferation and promoting apoptosis. Genes Dev. 2003, 17, 2514–2519. [Google Scholar] [CrossRef]
  4. Justice, R.W.; Zilian, O.; Woods, D.F.; Noll, M.; Bryant, P.J. The Drosophila tumor suppressor gene warts encodes a homolog of human myotonic dystrophy kinase and is required for the control of cell shape and proliferation. Genes Dev. 1995, 9, 534–546. [Google Scholar] [CrossRef]
  5. Pantalacci, S.; Tapon, N.; Leopold, P. The Salvador partner Hippo promotes apoptosis and cell-cycle exit in Drosophila. Nat. Cell Biol. 2003, 5, 921–927. [Google Scholar] [CrossRef] [PubMed]
  6. Xu, T.; Wang, W.; Zhang, S.; Stewart, R.A.; Yu, W. Identifying tumor suppressors in genetic mosaics: The Drosophila lats gene encodes a putative protein kinase. Development 1995, 121, 1053–1063. [Google Scholar] [PubMed]
  7. Udan, R.S.; Kango-Singh, M.; Nolo, R.; Tao, C.; Halder, G. Hippo promotes proliferation arrest and apoptosis in the Salvador/Warts pathway. Nat. Cell Biol. 2003, 5, 914–920. [Google Scholar] [CrossRef] [PubMed]
  8. Wu, S.; Huang, J.; Dong, J.; Pan, D. hippo encodes a Ste-20 family protein kinase that restricts cell proliferation and promotes apoptosis in conjunction with salvador and warts. Cell 2003, 114, 445–456. [Google Scholar] [CrossRef]
  9. Kango-Singh, M.; Nolo, R.; Tao, C.; Verstreken, P.; Hiesinger, P.R.; Bellen, H.J.; Halder, G. Shar-pei mediates cell proliferation arrest during imaginal disc growth in Drosophila. Development 2002, 129, 5719–5730. [Google Scholar] [CrossRef] [PubMed]
  10. Tapon, N.; Harvey, K.F.; Bell, D.W.; Wahrer, D.C.; Schiripo, T.A.; Haber, D.; Hariharan, I.K. salvador Promotes both cell cycle exit and apoptosis in Drosophila and is mutated in human cancer cell lines. Cell 2002, 110, 467–478. [Google Scholar] [CrossRef]
  11. Lai, Z.C.; Wei, X.; Shimizu, T.; Ramos, E.; Rohrbaugh, M.; Nikolaidis, N.; Ho, L.L.; Li, Y. Control of cell proliferation and apoptosis by mob as tumor suppressor, mats. Cell 2005, 120, 675–685. [Google Scholar] [CrossRef] [PubMed]
  12. Ren, F.; Zhang, L.; Jiang, J. Hippo signaling regulates Yorkie nuclear localization and activity through 14-3-3 dependent and independent mechanisms. Dev. Biol. 2010, 337, 303–312. [Google Scholar] [CrossRef]
  13. Huang, J.; Wu, S.; Barrera, J.; Matthews, K.; Pan, D. The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila Homolog of YAP. Cell 2005, 122, 421–434. [Google Scholar] [CrossRef] [PubMed]
  14. Dong, J.; Feldmann, G.; Huang, J.; Wu, S.; Zhang, N.; Comerford, S.A.; Gayyed, M.F.; Anders, R.A.; Maitra, A.; Pan, D. Elucidation of a universal size-control mechanism in Drosophila and mammals. Cell 2007, 130, 1120–1133. [Google Scholar] [CrossRef] [PubMed]
  15. Oh, H.; Irvine, K.D. In vivo regulation of Yorkie phosphorylation and localization. Development 2008, 135, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
  16. Chan, E.H.; Nousiainen, M.; Chalamalasetty, R.B.; Schafer, A.; Nigg, E.A.; Sillje, H.H. The Ste20-like kinase Mst2 activates the human large tumor suppressor kinase Lats1. Oncogene 2005, 24, 2076–2086. [Google Scholar] [CrossRef]
  17. Praskova, M.; Xia, F.; Avruch, J. MOBKL1A/MOBKL1B phosphorylation by MST1 and MST2 inhibits cell proliferation. Curr. Biol. 2008, 18, 311–321. [Google Scholar] [CrossRef]
  18. Callus, B.A.; Verhagen, A.M.; Vaux, D.L. Association of mammalian sterile twenty kinases, Mst1 and Mst2, with hSalvador via C-terminal coiled-coil domains, leads to its stabilization and phosphorylation. FEBS J. 2006, 273, 4264–4276. [Google Scholar] [CrossRef] [PubMed]
  19. Staley, B.K.; Irvine, K.D. Hippo signaling in Drosophila: Recent advances and insights. Dev. Dyn. 2012, 241, 3–15. [Google Scholar] [CrossRef]
  20. Sudol, M. Yes-associated protein (YAP65) is a proline-rich phosphoprotein that binds to the SH3 domain of the Yes proto-oncogene product. Oncogene 1994, 9, 2145–2152. [Google Scholar]
  21. Kanai, F.; Marignani, P.A.; Sarbassova, D.; Yagi, R.; Hall, R.A.; Donowitz, M.; Hisaminato, A.; Fujiwara, T.; Ito, Y.; Cantley, L.C.; et al. TAZ: A novel transcriptional co-activator regulated by interactions with 14-3-3 and PDZ domain proteins. EMBO J. 2000, 19, 6778–6791. [Google Scholar] [CrossRef]
  22. Zhao, B.; Wei, X.; Li, W.; Udan, R.S.; Yang, Q.; Kim, J.; Xie, J.; Ikenoue, T.; Yu, J.; Li, L.; et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 2007, 21, 2747–2761. [Google Scholar] [CrossRef]
  23. Hao, Y.; Chun, A.; Cheung, K.; Rashidi, B.; Yang, X. Tumor suppressor LATS1 is a negative regulator of oncogene YAP. J. Biol. Chem. 2008, 283, 5496–5509. [Google Scholar] [CrossRef] [PubMed]
  24. Oka, T.; Mazack, V.; Sudol, M. Mst2 and Lats kinases regulate apoptotic function of Yes kinase-associated protein (YAP). J. Biol. Chem. 2008, 283, 27534–27546. [Google Scholar] [CrossRef] [PubMed]
  25. Lei, Q.Y.; Zhang, H.; Zhao, B.; Zha, Z.Y.; Bai, F.; Pei, X.H.; Zhao, S.; Xiong, Y.; Guan, K.L. TAZ promotes cell proliferation and epithelial-mesenchymal transition and is inhibited by the hippo pathway. Mol. Cell. Biol. 2008, 28, 2426–2436. [Google Scholar] [CrossRef]
  26. Liu, C.Y.; Zha, Z.Y.; Zhou, X.; Zhang, H.; Huang, W.; Zhao, D.; Li, T.; Chan, S.W.; Lim, C.J.; Hong, W.; et al. The hippo tumor pathway promotes TAZ degradation by phosphorylating a phosphodegron and recruiting the SCF{beta}-TrCP E3 ligase. J. Biol. Chem. 2010, 285, 37159–37169. [Google Scholar] [CrossRef] [PubMed]
  27. Zhao, B.; Ye, X.; Yu, J.; Li, L.; Li, W.; Li, S.; Yu, J.; Lin, J.D.; Wang, C.Y.; Chinnaiyan, A.M.; et al. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev. 2008, 22, 1962–1971. [Google Scholar] [CrossRef]
  28. Zhang, H.; Liu, C.Y.; Zha, Z.Y.; Zhao, B.; Yao, J.; Zhao, S.; Xiong, Y.; Lei, Q.Y.; Guan, K.L. TEAD transcription factors mediate the function of TAZ in cell growth and epithelial-mesenchymal transition. J. Biol. Chem. 2009, 284, 13355–13362. [Google Scholar] [CrossRef] [PubMed]
  29. Meng, Z.; Moroishi, T.; Guan, K.L. Mechanisms of Hippo pathway regulation. Genes Dev. 2016, 30, 1–17. [Google Scholar] [CrossRef] [PubMed]
  30. Zanconato, F.; Cordenonsi, M.; Piccolo, S. YAP/TAZ at the Roots of Cancer. Cancer Cell 2016, 29, 783–803. [Google Scholar] [CrossRef]
  31. Mo, J.S.; Park, H.W.; Guan, K.L. The Hippo signaling pathway in stem cell biology and cancer. EMBO Rep. 2014, 15, 642–656. [Google Scholar] [CrossRef]
  32. Mo, J.S. The role of extracellular biophysical cues in modulating the Hippo-YAP pathway. BMB Rep. 2017, 50, 71–78. [Google Scholar] [CrossRef]
  33. Corvol, H.; Flamein, F.; Epaud, R.; Clement, A.; Guillot, L. Lung alveolar epithelium and interstitial lung disease. Int. J. Biochem. Cell Biol. 2009, 41, 1643–1651. [Google Scholar] [CrossRef]
  34. Herriges, M.; Morrisey, E.E. Lung development: Orchestrating the generation and regeneration of a complex organ. Development 2014, 141, 502–513. [Google Scholar] [CrossRef] [PubMed]
  35. Lange, A.W.; Sridharan, A.; Xu, Y.; Stripp, B.R.; Perl, A.K.; Whitsett, J.A. Hippo/Yap signaling controls epithelial progenitor cell proliferation and differentiation in the embryonic and adult lung. J. Mol. Cell Biol. 2015, 7, 35–47. [Google Scholar] [CrossRef]
  36. Zhao, R.; Fallon, T.R.; Saladi, S.V.; Pardo-Saganta, A.; Villoria, J.; Mou, H.; Vinarsky, V.; Gonzalez-Celeiro, M.; Nunna, N.; Hariri, L.P.; et al. Yap tunes airway epithelial size and architecture by regulating the identity, maintenance, and self-renewal of stem cells. Dev. Cell 2014, 30, 151–165. [Google Scholar] [CrossRef] [PubMed]
  37. Park, K.S.; Whitsett, J.A.; Di Palma, T.; Hong, J.H.; Yaffe, M.B.; Zannini, M. TAZ interacts with TTF-1 and regulates expression of surfactant protein-C. J. Biol. Chem. 2004, 279, 17384–17390. [Google Scholar] [CrossRef]
  38. Mitani, A.; Nagase, T.; Fukuchi, K.; Aburatani, H.; Makita, R.; Kurihara, H. Transcriptional coactivator with PDZ-binding motif is essential for normal alveolarization in mice. Am. J. Respir. Crit. Care Med. 2009, 180, 326–338. [Google Scholar] [CrossRef] [PubMed]
  39. Kasai, H.; Allen, J.T.; Mason, R.M.; Kamimura, T.; Zhang, Z. TGF-beta1 induces human alveolar epithelial to mesenchymal cell transition (EMT). Respir. Res. 2005, 6, 56. [Google Scholar] [CrossRef]
  40. Lasky, J.A.; Ortiz, L.A.; Tonthat, B.; Hoyle, G.W.; Corti, M.; Athas, G.; Lungarella, G.; Brody, A.; Friedman, M. Connective tissue growth factor mRNA expression is upregulated in bleomycin-induced lung fibrosis. Am. J. Physiol. 1998, 275, L365–L371. [Google Scholar] [CrossRef]
  41. Pan, L.H.; Yamauchi, K.; Uzuki, M.; Nakanishi, T.; Takigawa, M.; Inoue, H.; Sawai, T. Type II alveolar epithelial cells and interstitial fibroblasts express connective tissue growth factor in IPF. Eur. Respir. J. 2001, 17, 1220–1227. [Google Scholar] [CrossRef]
  42. Liu, F.; Lagares, D.; Choi, K.M.; Stopfer, L.; Marinkovic, A.; Vrbanac, V.; Probst, C.K.; Hiemer, S.E.; Sisson, T.H.; Horowitz, J.C.; et al. Mechanosignaling through YAP and TAZ drives fibroblast activation and fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 308, L344–L357. [Google Scholar] [CrossRef]
  43. Noguchi, S.; Saito, A.; Mikami, Y.; Urushiyama, H.; Horie, M.; Matsuzaki, H.; Takeshima, H.; Makita, K.; Miyashita, N.; Mitani, A.; et al. TAZ contributes to pulmonary fibrosis by activating profibrotic functions of lung fibroblasts. Sci. Rep. 2017, 7, 42595. [Google Scholar] [CrossRef]
  44. Jorgenson, A.J.; Choi, K.M.; Sicard, D.; Smith, K.M.; Hiemer, S.E.; Varelas, X.; Tschumperlin, D.J. TAZ activation drives fibroblast spheroid growth, expression of profibrotic paracrine signals, and context-dependent ECM gene expression. Am. J. Physiol. Cell Physiol. 2017, 312, C277–C285. [Google Scholar] [CrossRef]
  45. Chen, Y.; Zhao, X.; Sun, J.; Su, W.; Zhang, L.; Li, Y.; Liu, Y.; Zhang, L.; Lu, Y.; Shan, H.; et al. YAP1/Twist promotes fibroblast activation and lung fibrosis that conferred by miR-15a loss in IPF. Cell Death Differ. 2019. [Google Scholar] [CrossRef]
  46. Swigris, J.J.; Gould, M.K.; Wilson, S.R. Health-related quality of life among patients with idiopathic pulmonary fibrosis. Chest 2005, 127, 284–294. [Google Scholar] [CrossRef] [PubMed]
  47. Katzenstein, A.L.; Myers, J.L. Idiopathic pulmonary fibrosis: Clinical relevance of pathologic classification. Am. J. Respir. Crit. Care Med. 1998, 157, 1301–1315. [Google Scholar] [CrossRef] [PubMed]
  48. Xu, Y.; Mizuno, T.; Sridharan, A.; Du, Y.; Guo, M.; Tang, J.; Wikenheiser-Brokamp, K.A.; Perl, A.T.; Funari, V.A.; Gokey, J.J.; et al. Single-cell RNA sequencing identifies diverse roles of epithelial cells in idiopathic pulmonary fibrosis. JCI Insight 2016, 1, e90558. [Google Scholar] [CrossRef]
  49. Gokey, J.J.; Sridharan, A.; Xu, Y.; Green, J.; Carraro, G.; Stripp, B.R.; Perl, A.T.; Whitsett, J.A. Active epithelial Hippo signaling in idiopathic pulmonary fibrosis. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed]
  50. Speight, P.; Kofler, M.; Szaszi, K.; Kapus, A. Context-dependent switch in chemo/mechanotransduction via multilevel crosstalk among cytoskeleton-regulated MRTF and TAZ and TGFbeta-regulated Smad3. Nat. Commun. 2016, 7, 11642. [Google Scholar] [CrossRef]
  51. Miner, J.H. The glomerular basement membrane. Exp. Cell Res. 2012, 318, 973–978. [Google Scholar] [CrossRef] [PubMed]
  52. Hossain, Z.; Ali, S.M.; Ko, H.L.; Xu, J.; Ng, C.P.; Guo, K.; Qi, Z.; Ponniah, S.; Hong, W.; Hunziker, W. Glomerulocystic kidney disease in mice with a targeted inactivation of Wwtr1. Proc. Natl. Acad. Sci. USA 2007, 104, 1631–1636. [Google Scholar] [CrossRef]
  53. Reginensi, A.; Scott, R.P.; Gregorieff, A.; Bagherie-Lachidan, M.; Chung, C.; Lim, D.S.; Pawson, T.; Wrana, J.; McNeill, H. Yap- and Cdc42-dependent nephrogenesis and morphogenesis during mouse kidney development. PLoS Genet. 2013, 9, e1003380. [Google Scholar] [CrossRef] [PubMed]
  54. Tian, Y.; Kolb, R.; Hong, J.H.; Carroll, J.; Li, D.; You, J.; Bronson, R.; Yaffe, M.B.; Zhou, J.; Benjamin, T. TAZ promotes PC2 degradation through a SCFbeta-Trcp E3 ligase complex. Mol. Cell. Biol. 2007, 27, 6383–6395. [Google Scholar] [CrossRef]
  55. Habbig, S.; Bartram, M.P.; Sagmuller, J.G.; Griessmann, A.; Franke, M.; Muller, R.U.; Schwarz, R.; Hoehne, M.; Bergmann, C.; Tessmer, C.; et al. The ciliopathy disease protein NPHP9 promotes nuclear delivery and activation of the oncogenic transcriptional regulator TAZ. Hum. Mol. Genet. 2012, 21, 5528–5538. [Google Scholar] [CrossRef] [PubMed]
  56. Habbig, S.; Bartram, M.P.; Muller, R.U.; Schwarz, R.; Andriopoulos, N.; Chen, S.; Sagmuller, J.G.; Hoehne, M.; Burst, V.; Liebau, M.C.; et al. NPHP4, a cilia-associated protein, negatively regulates the Hippo pathway. J. Cell Biol. 2011, 193, 633–642. [Google Scholar] [CrossRef]
  57. Happe, H.; van der Wal, A.M.; Leonhard, W.N.; Kunnen, S.J.; Breuning, M.H.; de Heer, E.; Peters, D.J. Altered Hippo signalling in polycystic kidney disease. J. Pathol. 2011, 224, 133–142. [Google Scholar] [CrossRef]
  58. Seo, E.; Kim, W.Y.; Hur, J.; Kim, H.; Nam, S.A.; Choi, A.; Kim, Y.M.; Park, S.H.; Chung, C.; Kim, J.; et al. The Hippo-Salvador signaling pathway regulates renal tubulointerstitial fibrosis. Sci. Rep. 2016, 6, 31931. [Google Scholar] [CrossRef]
  59. Anorga, S.; Overstreet, J.M.; Falke, L.L.; Tang, J.; Goldschmeding, R.G.; Higgins, P.J.; Samarakoon, R. Deregulation of Hippo-TAZ pathway during renal injury confers a fibrotic maladaptive phenotype. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2018, 32, 2644–2657. [Google Scholar] [CrossRef]
  60. Xu, J.; Li, P.X.; Wu, J.; Gao, Y.J.; Yin, M.X.; Lin, Y.; Yang, M.; Chen, D.P.; Sun, H.P.; Liu, Z.B.; et al. Involvement of the Hippo pathway in regeneration and fibrogenesis after ischaemic acute kidney injury: YAP is the key effector. Clin. Sci. 2016, 130, 349–363. [Google Scholar] [CrossRef] [PubMed]
  61. Wennmann, D.O.; Vollenbroker, B.; Eckart, A.K.; Bonse, J.; Erdmann, F.; Wolters, D.A.; Schenk, L.K.; Schulze, U.; Kremerskothen, J.; Weide, T.; et al. The Hippo pathway is controlled by Angiotensin II signaling and its reactivation induces apoptosis in podocytes. Cell Death Dis. 2014, 5, e1519. [Google Scholar] [CrossRef]
  62. Rinschen, M.M.; Grahammer, F.; Hoppe, A.K.; Kohli, P.; Hagmann, H.; Kretz, O.; Bertsch, S.; Hohne, M.; Gobel, H.; Bartram, M.P.; et al. YAP-mediated mechanotransduction determines the podocyte’s response to damage. Sci. Signal. 2017, 10, eaaf8165. [Google Scholar] [CrossRef]
  63. Schwartzman, M.; Reginensi, A.; Wong, J.S.; Basgen, J.M.; Meliambro, K.; Nicholas, S.B.; D’Agati, V.; McNeill, H.; Campbell, K.N. Podocyte-Specific Deletion of Yes-Associated Protein Causes FSGS and Progressive Renal Failure. J. Am. Soc. Nephrol. JASN 2016, 27, 216–226. [Google Scholar] [CrossRef] [PubMed]
  64. Szeto, S.G.; Narimatsu, M.; Lu, M.; He, X.; Sidiqi, A.M.; Tolosa, M.F.; Chan, L.; De Freitas, K.; Bialik, J.F.; Majumder, S.; et al. YAP/TAZ Are Mechanoregulators of TGF-beta-Smad Signaling and Renal Fibrogenesis. J. Am. Soc. Nephrol. JASN 2016, 27, 3117–3128. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, C.; Zhang, L.; He, Q.; Feng, X.; Zhu, J.; Xu, Z.; Wang, X.; Chen, F.; Li, X.; Dong, J. Differences in Yes-associated protein and mRNA levels in regenerating liver and hepatocellular carcinoma. Mol. Med. Rep. 2012, 5, 410–414. [Google Scholar] [CrossRef]
  66. Camargo, F.D.; Gokhale, S.; Johnnidis, J.B.; Fu, D.; Bell, G.W.; Jaenisch, R.; Brummelkamp, T.R. YAP1 increases organ size and expands undifferentiated progenitor cells. Curr. Biol. 2007, 17, 2054–2060. [Google Scholar] [CrossRef]
  67. Zhou, D.; Conrad, C.; Xia, F.; Park, J.S.; Payer, B.; Yin, Y.; Lauwers, G.Y.; Thasler, W.; Lee, J.T.; Avruch, J.; et al. Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell 2009, 16, 425–438. [Google Scholar] [CrossRef]
  68. Lu, L.; Li, Y.; Kim, S.M.; Bossuyt, W.; Liu, P.; Qiu, Q.; Wang, Y.; Halder, G.; Finegold, M.J.; Lee, J.S.; et al. Hippo signaling is a potent in vivo growth and tumor suppressor pathway in the mammalian liver. Proc. Natl. Acad. Sci. USA 2010, 107, 1437–1442. [Google Scholar] [CrossRef]
  69. Mannaerts, I.; Leite, S.B.; Verhulst, S.; Claerhout, S.; Eysackers, N.; Thoen, L.F.; Hoorens, A.; Reynaert, H.; Halder, G.; van Grunsven, L.A. The Hippo pathway effector YAP controls mouse hepatic stellate cell activation. J. Hepatol. 2015, 63, 679–688. [Google Scholar] [CrossRef]
  70. Du, K.; Hyun, J.; Premont, R.T.; Choi, S.S.; Michelotti, G.A.; Swiderska-Syn, M.; Dalton, G.D.; Thelen, E.; Rizi, B.S.; Jung, Y.; et al. Hedgehog-YAP Signaling Pathway Regulates Glutaminolysis to Control Activation of Hepatic Stellate Cells. Gastroenterology 2018, 154, 1465.e13–1479.e13. [Google Scholar] [CrossRef] [PubMed]
  71. Wang, X.; Zheng, Z.; Caviglia, J.M.; Corey, K.E.; Herfel, T.M.; Cai, B.; Masia, R.; Chung, R.T.; Lefkowitch, J.H.; Schwabe, R.F.; et al. Hepatocyte TAZ/WWTR1 Promotes Inflammation and Fibrosis in Nonalcoholic Steatohepatitis. Cell Metab. 2016, 24, 848–862. [Google Scholar] [CrossRef] [PubMed]
  72. Laflamme, M.A.; Murry, C.E. Heart regeneration. Nature 2011, 473, 326–335. [Google Scholar] [CrossRef] [PubMed]
  73. Heallen, T.; Zhang, M.; Wang, J.; Bonilla-Claudio, M.; Klysik, E.; Johnson, R.L.; Martin, J.F. Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science 2011, 332, 458–461. [Google Scholar] [CrossRef] [PubMed]
  74. Heallen, T.; Morikawa, Y.; Leach, J.; Tao, G.; Willerson, J.T.; Johnson, R.L.; Martin, J.F. Hippo signaling impedes adult heart regeneration. Development 2013, 140, 4683–4690. [Google Scholar] [CrossRef] [PubMed]
  75. Xin, M.; Kim, Y.; Sutherland, L.B.; Qi, X.; McAnally, J.; Schwartz, R.J.; Richardson, J.A.; Bassel-Duby, R.; Olson, E.N. Regulation of insulin-like growth factor signaling by Yap governs cardiomyocyte proliferation and embryonic heart size. Sci. Signal. 2011, 4, ra70. [Google Scholar] [CrossRef]
  76. Von Gise, A.; Lin, Z.; Schlegelmilch, K.; Honor, L.B.; Pan, G.M.; Buck, J.N.; Ma, Q.; Ishiwata, T.; Zhou, B.; Camargo, F.D.; et al. YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy. Proc. Natl. Acad. Sci. USA 2012, 109, 2394–2399. [Google Scholar] [CrossRef]
  77. Xin, M.; Kim, Y.; Sutherland, L.B.; Murakami, M.; Qi, X.; McAnally, J.; Porrello, E.R.; Mahmoud, A.I.; Tan, W.; Shelton, J.M.; et al. Hippo pathway effector Yap promotes cardiac regeneration. Proc. Natl. Acad. Sci. USA 2013, 110, 13839–13844. [Google Scholar] [CrossRef]
  78. Del Re, D.P.; Yang, Y.; Nakano, N.; Cho, J.; Zhai, P.; Yamamoto, T.; Zhang, N.; Yabuta, N.; Nojima, H.; Pan, D.; et al. Yes-associated protein isoform 1 (Yap1) promotes cardiomyocyte survival and growth to protect against myocardial ischemic injury. J. Biol. Chem. 2013, 288, 3977–3988. [Google Scholar] [CrossRef]
  79. Wu, P.; Liu, Z.; Zhao, T.; Xia, F.; Gong, L.; Zheng, Z.; Chen, Z.; Yang, T.; Duan, Q. Lovastatin attenuates angiotensin II induced cardiovascular fibrosis through the suppression of YAP/TAZ signaling. Biochem. Biophys. Res. Commun. 2019, 512, 736–741. [Google Scholar] [CrossRef]
  80. Schlegelmilch, K.; Mohseni, M.; Kirak, O.; Pruszak, J.; Rodriguez, J.R.; Zhou, D.; Kreger, B.T.; Vasioukhin, V.; Avruch, J.; Brummelkamp, T.R.; et al. Yap1 acts downstream of alpha-catenin to control epidermal proliferation. Cell 2011, 144, 782–795. [Google Scholar] [CrossRef]
  81. Rognoni, E.; Walko, G. The Roles of YAP/TAZ and the Hippo Pathway in Healthy and Diseased Skin. Cells 2019, 8, 411. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, H.; Pasolli, H.A.; Fuchs, E. Yes-associated protein (YAP) transcriptional coactivator functions in balancing growth and differentiation in skin. Proc. Natl. Acad. Sci. USA 2011, 108, 2270–2275. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, M.J.; Byun, M.R.; Furutani-Seiki, M.; Hong, J.H.; Jung, H.S. YAP and TAZ regulate skin wound healing. J. Investig. Dermatol. 2014, 134, 518–525. [Google Scholar] [CrossRef] [PubMed]
  84. Piersma, B.; de Rond, S.; Werker, P.M.; Boo, S.; Hinz, B.; van Beuge, M.M.; Bank, R.A. YAP1 Is a Driver of Myofibroblast Differentiation in Normal and Diseased Fibroblasts. Am. J. Pathol. 2015, 185, 3326–3337. [Google Scholar] [CrossRef]
  85. Toyama, T.; Looney, A.P.; Baker, B.M.; Stawski, L.; Haines, P.; Simms, R.; Szymaniak, A.D.; Varelas, X.; Trojanowska, M. Therapeutic Targeting of TAZ and YAP by Dimethyl Fumarate in Systemic Sclerosis Fibrosis. J. Investig. Dermatol. 2018, 138, 78–88. [Google Scholar] [CrossRef]
  86. Darby, I.A.; Zakuan, N.; Billet, F.; Desmouliere, A. The myofibroblast, a key cell in normal and pathological tissue repair. Cell. Mol. Life Sci. 2016, 73, 1145–1157. [Google Scholar] [CrossRef] [PubMed]
  87. Nieman, K.M.; Kenny, H.A.; Penicka, C.V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M.R.; Romero, I.L.; Carey, M.S.; Mills, G.B.; Hotamisligil, G.S.; et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011, 17, 1498–1503. [Google Scholar] [CrossRef]
  88. Lu, P.; Weaver, V.M.; Werb, Z. The extracellular matrix: A dynamic niche in cancer progression. J. Cell Biol. 2012, 196, 395–406. [Google Scholar] [CrossRef]
  89. Mueller, M.M.; Fusenig, N.E. Friends or foes—Bipolar effects of the tumour stroma in cancer. Nat. Rev. Cancer 2004, 4, 839–849. [Google Scholar] [CrossRef]
  90. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
  91. Kalluri, R.; Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 2006, 6, 392–401. [Google Scholar] [CrossRef]
  92. Gaggioli, C.; Hooper, S.; Hidalgo-Carcedo, C.; Grosse, R.; Marshall, J.F.; Harrington, K.; Sahai, E. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat. Cell Biol. 2007, 9, 1392–1400. [Google Scholar] [CrossRef]
  93. Glentis, A.; Oertle, P.; Mariani, P.; Chikina, A.; El Marjou, F.; Attieh, Y.; Zaccarini, F.; Lae, M.; Loew, D.; Dingli, F.; et al. Cancer-associated fibroblasts induce metalloprotease-independent cancer cell invasion of the basement membrane. Nat. Commun. 2017, 8, 924. [Google Scholar] [CrossRef] [PubMed]
  94. Calvo, F.; Ege, N.; Grande-Garcia, A.; Hooper, S.; Jenkins, R.P.; Chaudhry, S.I.; Harrington, K.; Williamson, P.; Moeendarbary, E.; Charras, G.; et al. Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat. Cell Biol. 2013, 15, 637–646. [Google Scholar] [CrossRef] [PubMed]
  95. Chan, S.W.; Lim, C.J.; Guo, K.; Ng, C.P.; Lee, I.; Hunziker, W.; Zeng, Q.; Hong, W. A role for TAZ in migration, invasion, and tumorigenesis of breast cancer cells. Cancer Res. 2008, 68, 2592–2598. [Google Scholar] [CrossRef]
  96. Foster, C.T.; Gualdrini, F.; Treisman, R. Mutual dependence of the MRTF-SRF and YAP-TEAD pathways in cancer-associated fibroblasts is indirect and mediated by cytoskeletal dynamics. Genes Dev. 2017, 31, 2361–2375. [Google Scholar] [CrossRef] [PubMed]
  97. Yazdani, S.; Bansal, R.; Prakash, J. Drug targeting to myofibroblasts: Implications for fibrosis and cancer. Adv. Drug Deliv. Rev. 2017, 121, 101–116. [Google Scholar] [CrossRef] [PubMed]
  98. Cernaro, V.; Lacquaniti, A.; Donato, V.; Fazio, M.R.; Buemi, A.; Buemi, M. Fibrosis, regeneration and cancer: What is the link? Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. Eur. Ren. Assoc. 2012, 27, 21–27. [Google Scholar] [CrossRef] [PubMed]
  99. Mehner, C.; Radisky, D.C. Triggering the landslide: The tumor-promotional effects of myofibroblasts. Exp. Cell Res. 2013, 319, 1657–1662. [Google Scholar] [CrossRef]
  100. Otranto, M.; Sarrazy, V.; Bonte, F.; Hinz, B.; Gabbiani, G.; Desmouliere, A. The role of the myofibroblast in tumor stroma remodeling. Cell Adhes. Migr. 2012, 6, 203–219. [Google Scholar] [CrossRef] [PubMed]
  101. Pan, D. The hippo signaling pathway in development and cancer. Dev. Cell 2010, 19, 491–505. [Google Scholar] [CrossRef] [PubMed]
  102. Plouffe, S.W.; Hong, A.W.; Guan, K.L. Disease implications of the Hippo/YAP pathway. Trends Mol. Med. 2015, 21, 212–222. [Google Scholar] [CrossRef] [PubMed]
  103. Bueno, R.; Stawiski, E.W.; Goldstein, L.D.; Durinck, S.; De Rienzo, A.; Modrusan, Z.; Gnad, F.; Nguyen, T.T.; Jaiswal, B.S.; Chirieac, L.R.; et al. Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations. Nat. Genet. 2016, 48, 407–416. [Google Scholar] [CrossRef] [PubMed]
  104. Oh, J.E.; Ohta, T.; Satomi, K.; Foll, M.; Durand, G.; McKay, J.; Le Calvez-Kelm, F.; Mittelbronn, M.; Brokinkel, B.; Paulus, W.; et al. Alterations in the NF2/LATS1/LATS2/YAP Pathway in Schwannomas. J. Neuropathol. Exp. Neurol. 2015, 74, 952–959. [Google Scholar] [CrossRef] [PubMed]
  105. Evans, D.G. Neurofibromatosis 2 [Bilateral acoustic neurofibromatosis, central neurofibromatosis, NF2, neurofibromatosis type II]. Genet. Med. Off. J. Am. Coll. Med Genet. 2009, 11, 599–610. [Google Scholar] [CrossRef] [PubMed]
  106. Endoh, H.; Yatabe, Y.; Shimizu, S.; Tajima, K.; Kuwano, H.; Takahashi, T.; Mitsudomi, T. RASSF1A gene inactivation in non-small cell lung cancer and its clinical implication. Int. J. Cancer 2003, 106, 45–51. [Google Scholar] [CrossRef]
  107. Takahashi, Y.; Miyoshi, Y.; Takahata, C.; Irahara, N.; Taguchi, T.; Tamaki, Y.; Noguchi, S. Down-regulation of LATS1 and LATS2 mRNA expression by promoter hypermethylation and its association with biologically aggressive phenotype in human breast cancers. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2005, 11, 1380–1385. [Google Scholar] [CrossRef] [PubMed]
  108. Seidel, C.; Schagdarsurengin, U.; Blumke, K.; Wurl, P.; Pfeifer, G.P.; Hauptmann, S.; Taubert, H.; Dammann, R. Frequent hypermethylation of MST1 and MST2 in soft tissue sarcoma. Mol. Carcinog. 2007, 46, 865–871. [Google Scholar] [CrossRef] [PubMed]
  109. Song, H.; Mak, K.K.; Topol, L.; Yun, K.; Hu, J.; Garrett, L.; Chen, Y.; Park, O.; Chang, J.; Simpson, R.M.; et al. Mammalian Mst1 and Mst2 kinases play essential roles in organ size control and tumor suppression. Proc. Natl. Acad. Sci. USA 2010, 107, 1431–1436. [Google Scholar] [CrossRef] [PubMed]
  110. Zhou, D.; Zhang, Y.; Wu, H.; Barry, E.; Yin, Y.; Lawrence, E.; Dawson, D.; Willis, J.E.; Markowitz, S.D.; Camargo, F.D.; et al. Mst1 and Mst2 protein kinases restrain intestinal stem cell proliferation and colonic tumorigenesis by inhibition of Yes-associated protein (Yap) overabundance. Proc. Natl. Acad. Sci. USA 2011, 108, E1312–E1320. [Google Scholar] [CrossRef] [PubMed]
  111. St John, M.A.; Tao, W.; Fei, X.; Fukumoto, R.; Carcangiu, M.L.; Brownstein, D.G.; Parlow, A.F.; McGrath, J.; Xu, T. Mice deficient of Lats1 develop soft-tissue sarcomas, ovarian tumours and pituitary dysfunction. Nat. Genet. 1999, 21, 182–186. [Google Scholar] [CrossRef] [PubMed]
  112. Jiang, Z.; Li, X.; Hu, J.; Zhou, W.; Jiang, Y.; Li, G.; Lu, D. Promoter hypermethylation-mediated down-regulation of LATS1 and LATS2 in human astrocytoma. Neurosci. Res. 2006, 56, 450–458. [Google Scholar] [CrossRef] [PubMed]
  113. Zender, L.; Spector, M.S.; Xue, W.; Flemming, P.; Cordon-Cardo, C.; Silke, J.; Fan, S.T.; Luk, J.M.; Wigler, M.; Hannon, G.J.; et al. Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell 2006, 125, 1253–1267. [Google Scholar] [CrossRef] [PubMed]
  114. Overholtzer, M.; Zhang, J.; Smolen, G.A.; Muir, B.; Li, W.; Sgroi, D.C.; Deng, C.X.; Brugge, J.S.; Haber, D.A. Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc. Natl. Acad. Sci. USA 2006, 103, 12405–12410. [Google Scholar] [CrossRef] [PubMed]
  115. Tanas, M.R.; Sboner, A.; Oliveira, A.M.; Erickson-Johnson, M.R.; Hespelt, J.; Hanwright, P.J.; Flanagan, J.; Luo, Y.; Fenwick, K.; Natrajan, R.; et al. Identification of a disease-defining gene fusion in epithelioid hemangioendothelioma. Sci. Transl. Med. 2011, 3, 98ra82. [Google Scholar] [CrossRef]
  116. Antonescu, C.R.; Le Loarer, F.; Mosquera, J.M.; Sboner, A.; Zhang, L.; Chen, C.L.; Chen, H.W.; Pathan, N.; Krausz, T.; Dickson, B.C.; et al. Novel YAP1-TFE3 fusion defines a distinct subset of epithelioid hemangioendothelioma. Geneschromosomes Cancer 2013, 52, 775–784. [Google Scholar] [CrossRef]
  117. Ota, M.; Sasaki, H. Mammalian Tead proteins regulate cell proliferation and contact inhibition as transcriptional mediators of Hippo signaling. Development 2008, 135, 4059–4069. [Google Scholar] [CrossRef]
  118. Nishioka, N.; Inoue, K.; Adachi, K.; Kiyonari, H.; Ota, M.; Ralston, A.; Yabuta, N.; Hirahara, S.; Stephenson, R.O.; Ogonuki, N.; et al. The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev. Cell 2009, 16, 398–410. [Google Scholar] [CrossRef] [PubMed]
  119. Gumbiner, B.M.; Kim, N.G. The Hippo-YAP signaling pathway and contact inhibition of growth. J. Cell Sci. 2014, 127, 709–717. [Google Scholar] [CrossRef]
  120. Dupont, S.; Morsut, L.; Aragona, M.; Enzo, E.; Giulitti, S.; Cordenonsi, M.; Zanconato, F.; Le Digabel, J.; Forcato, M.; Bicciato, S.; et al. Role of YAP/TAZ in mechanotransduction. Nature 2011, 474, 179–183. [Google Scholar] [CrossRef] [PubMed]
  121. Zhao, B.; Li, L.; Wang, L.; Wang, C.Y.; Yu, J.; Guan, K.L. Cell detachment activates the Hippo pathway via cytoskeleton reorganization to induce anoikis. Genes Dev. 2012, 26, 54–68. [Google Scholar] [CrossRef] [PubMed]
  122. Driscoll, T.P.; Cosgrove, B.D.; Heo, S.J.; Shurden, Z.E.; Mauck, R.L. Cytoskeletal to Nuclear Strain Transfer Regulates YAP Signaling in Mesenchymal Stem Cells. Biophys. J. 2015, 108, 2783–2793. [Google Scholar] [CrossRef]
  123. Aragona, M.; Panciera, T.; Manfrin, A.; Giulitti, S.; Michielin, F.; Elvassore, N.; Dupont, S.; Piccolo, S. A mechanical checkpoint controls multicellular growth through YAP/TAZ regulation by actin-processing factors. Cell 2013, 154, 1047–1059. [Google Scholar] [CrossRef] [PubMed]
  124. Schwartz, M.A. Integrins and extracellular matrix in mechanotransduction. Cold Spring Harb. Perspect. Biol. 2010, 2, a005066. [Google Scholar] [CrossRef] [PubMed]
  125. Serrano, I.; McDonald, P.C.; Lock, F.; Muller, W.J.; Dedhar, S. Inactivation of the Hippo tumour suppressor pathway by integrin-linked kinase. Nat. Commun. 2013, 4, 2976. [Google Scholar] [CrossRef]
  126. Sabra, H.; Brunner, M.; Mandati, V.; Wehrle-Haller, B.; Lallemand, D.; Ribba, A.S.; Chevalier, G.; Guardiola, P.; Block, M.R.; Bouvard, D. beta1 integrin-dependent Rac/group I PAK signaling mediates YAP activation of Yes-associated protein 1 (YAP1) via NF2/merlin. J. Biol. Chem. 2017, 292, 19179–19197. [Google Scholar] [CrossRef]
  127. Kim, N.G.; Gumbiner, B.M. Adhesion to fibronectin regulates Hippo signaling via the FAK-Src-PI3K pathway. J. Cell Biol. 2015, 210, 503–515. [Google Scholar] [CrossRef] [PubMed]
  128. Nardone, G.; Oliver-De La Cruz, J.; Vrbsky, J.; Martini, C.; Pribyl, J.; Skladal, P.; Pesl, M.; Caluori, G.; Pagliari, S.; Martino, F.; et al. YAP regulates cell mechanics by controlling focal adhesion assembly. Nat. Commun. 2017, 8, 15321. [Google Scholar] [CrossRef]
  129. Wada, K.; Itoga, K.; Okano, T.; Yonemura, S.; Sasaki, H. Hippo pathway regulation by cell morphology and stress fibers. Development 2011, 138, 3907–3914. [Google Scholar] [CrossRef]
  130. Ikeda, S.; Cunningham, L.A.; Boggess, D.; Hawes, N.; Hobson, C.D.; Sundberg, J.P.; Naggert, J.K.; Smith, R.S.; Nishina, P.M. Aberrant actin cytoskeleton leads to accelerated proliferation of corneal epithelial cells in mice deficient for destrin (actin depolymerizing factor). Hum. Mol. Genet. 2003, 12, 1029–1037. [Google Scholar] [CrossRef]
  131. Chakraborty, S.; Lakshmanan, M.; Swa, H.L.; Chen, J.; Zhang, X.; Ong, Y.S.; Loo, L.S.; Akincilar, S.C.; Gunaratne, J.; Tergaonkar, V.; et al. An oncogenic role of Agrin in regulating focal adhesion integrity in hepatocellular carcinoma. Nat. Commun. 2015, 6, 6184. [Google Scholar] [CrossRef]
  132. Chakraborty, S.; Njah, K.; Pobbati, A.V.; Lim, Y.B.; Raju, A.; Lakshmanan, M.; Tergaonkar, V.; Lim, C.T.; Hong, W. Agrin as a Mechanotransduction Signal Regulating YAP through the Hippo Pathway. Cell Rep. 2017, 18, 2464–2479. [Google Scholar] [CrossRef]
  133. Chakraborty, S.; Hong, W. Linking Extracellular Matrix Agrin to the Hippo Pathway in Liver Cancer and Beyond. Cancers 2018, 10, 45. [Google Scholar] [CrossRef]
  134. Feng, M.; Gao, W.; Wang, R.; Chen, W.; Man, Y.G.; Figg, W.D.; Wang, X.W.; Dimitrov, D.S.; Ho, M. Therapeutically targeting glypican-3 via a conformation-specific single-domain antibody in hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2013, 110, E1083–E1091. [Google Scholar] [CrossRef]
  135. Kim, K.M.; Choi, Y.J.; Hwang, J.H.; Kim, A.R.; Cho, H.J.; Hwang, E.S.; Park, J.Y.; Lee, S.H.; Hong, J.H. Shear stress induced by an interstitial level of slow flow increases the osteogenic differentiation of mesenchymal stem cells through TAZ activation. PLoS ONE 2014, 9, e92427. [Google Scholar] [CrossRef]
  136. Wang, K.C.; Yeh, Y.T.; Nguyen, P.; Limqueco, E.; Lopez, J.; Thorossian, S.; Guan, K.L.; Li, Y.J.; Chien, S. Flow-dependent YAP/TAZ activities regulate endothelial phenotypes and atherosclerosis. Proc. Natl. Acad. Sci. USA 2016, 113, 11525–11530. [Google Scholar] [CrossRef] [PubMed]
  137. Sabine, A.; Bovay, E.; Demir, C.S.; Kimura, W.; Jaquet, M.; Agalarov, Y.; Zangger, N.; Scallan, J.P.; Graber, W.; Gulpinar, E.; et al. FOXC2 and fluid shear stress stabilize postnatal lymphatic vasculature. J. Clin. Investig. 2015, 125, 3861–3877. [Google Scholar] [CrossRef] [PubMed]
  138. Elosegui-Artola, A.; Andreu, I.; Beedle, A.E.M.; Lezamiz, A.; Uroz, M.; Kosmalska, A.J.; Oria, R.; Kechagia, J.Z.; Rico-Lastres, P.; Le Roux, A.L.; et al. Force Triggers YAP Nuclear Entry by Regulating Transport across Nuclear Pores. Cell 2017, 171, 1397.e1314–1410.e1314. [Google Scholar] [CrossRef] [PubMed]
  139. Sansores-Garcia, L.; Bossuyt, W.; Wada, K.; Yonemura, S.; Tao, C.; Sasaki, H.; Halder, G. Modulating F-actin organization induces organ growth by affecting the Hippo pathway. Embo J. 2011, 30, 2325–2335. [Google Scholar] [CrossRef]
  140. Fan, R.; Kim, N.G.; Gumbiner, B.M. Regulation of Hippo pathway by mitogenic growth factors via phosphoinositide 3-kinase and phosphoinositide-dependent kinase-1. Proc. Natl. Acad. Sci. USA 2013, 110, 2569–2574. [Google Scholar] [CrossRef]
  141. Wang, X.; Freire Valls, A.; Schermann, G.; Shen, Y.; Moya, I.M.; Castro, L.; Urban, S.; Solecki, G.M.; Winkler, F.; Riedemann, L.; et al. YAP/TAZ Orchestrate VEGF Signaling during Developmental Angiogenesis. Dev. Cell 2017, 42, 462–478. [Google Scholar] [CrossRef]
  142. Rizvi, S.; Yamada, D.; Hirsova, P.; Bronk, S.F.; Werneburg, N.W.; Krishnan, A.; Salim, W.; Zhang, L.; Trushina, E.; Truty, M.J.; et al. A Hippo and Fibroblast Growth Factor Receptor Autocrine Pathway in Cholangiocarcinoma. J. Biol. Chem. 2016, 291, 8031–8047. [Google Scholar] [CrossRef]
  143. Kapoor, A.; Yao, W.; Ying, H.; Hua, S.; Liewen, A.; Wang, Q.; Zhong, Y.; Wu, C.J.; Sadanandam, A.; Hu, B.; et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 2014, 158, 185–197. [Google Scholar] [CrossRef] [PubMed]
  144. Jang, W.; Kim, T.; Koo, J.S.; Kim, S.K.; Lim, D.S. Mechanical cue-induced YAP instructs Skp2-dependent cell cycle exit and oncogenic signaling. EMBO J. 2017, 36, 2510–2528. [Google Scholar] [CrossRef] [PubMed]
  145. Di Agostino, S.; Sorrentino, G.; Ingallina, E.; Valenti, F.; Ferraiuolo, M.; Bicciato, S.; Piazza, S.; Strano, S.; Del Sal, G.; Blandino, G. YAP enhances the pro-proliferative transcriptional activity of mutant p53 proteins. EMBO Rep. 2016, 17, 188–201. [Google Scholar] [CrossRef] [PubMed]
  146. Mizuno, T.; Murakami, H.; Fujii, M.; Ishiguro, F.; Tanaka, I.; Kondo, Y.; Akatsuka, S.; Toyokuni, S.; Yokoi, K.; Osada, H.; et al. YAP induces malignant mesothelioma cell proliferation by upregulating transcription of cell cycle-promoting genes. Oncogene 2012, 31, 5117–5122. [Google Scholar] [CrossRef] [PubMed]
  147. Zanconato, F.; Forcato, M.; Battilana, G.; Azzolin, L.; Quaranta, E.; Bodega, B.; Rosato, A.; Bicciato, S.; Cordenonsi, M.; Piccolo, S. Genome-wide association between YAP/TAZ/TEAD and AP-1 at enhancers drives oncogenic growth. Nat. Cell Biol. 2015, 17, 1218–1227. [Google Scholar] [CrossRef]
  148. Miyazono, K.; Katsuno, Y.; Koinuma, D.; Ehata, S.; Morikawa, M. Intracellular and extracellular TGF-beta signaling in cancer: Some recent topics. Front. Med. 2018, 12, 387–411. [Google Scholar] [CrossRef]
  149. Bora-Singhal, N.; Nguyen, J.; Schaal, C.; Perumal, D.; Singh, S.; Coppola, D.; Chellappan, S. YAP1 Regulates OCT4 Activity and SOX2 Expression to Facilitate Self-Renewal and Vascular Mimicry of Stem-Like Cells. Stem Cells 2015, 33, 1705–1718. [Google Scholar] [CrossRef] [PubMed]
  150. Shao, D.D.; Xue, W.; Krall, E.B.; Bhutkar, A.; Piccioni, F.; Wang, X.; Schinzel, A.C.; Sood, S.; Rosenbluh, J.; Kim, J.W.; et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 2014, 158, 171–184. [Google Scholar] [CrossRef]
  151. Cordenonsi, M.; Zanconato, F.; Azzolin, L.; Forcato, M.; Rosato, A.; Frasson, C.; Inui, M.; Montagner, M.; Parenti, A.R.; Poletti, A.; et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell 2011, 147, 759–772. [Google Scholar] [CrossRef]
  152. Lehmann, W.; Mossmann, D.; Kleemann, J.; Mock, K.; Meisinger, C.; Brummer, T.; Herr, R.; Brabletz, S.; Stemmler, M.P.; Brabletz, T. ZEB1 turns into a transcriptional activator by interacting with YAP1 in aggressive cancer types. Nat. Commun. 2016, 7, 10498. [Google Scholar] [CrossRef]
  153. Tang, Y.; Feinberg, T.; Keller, E.T.; Li, X.Y.; Weiss, S.J. Snail/Slug binding interactions with YAP/TAZ control skeletal stem cell self-renewal and differentiation. Nat. Cell Biol. 2016, 18, 917–929. [Google Scholar] [CrossRef]
  154. Bartucci, M.; Dattilo, R.; Moriconi, C.; Pagliuca, A.; Mottolese, M.; Federici, G.; Benedetto, A.D.; Todaro, M.; Stassi, G.; Sperati, F.; et al. TAZ is required for metastatic activity and chemoresistance of breast cancer stem cells. Oncogene 2015, 34, 681–690. [Google Scholar] [CrossRef] [PubMed]
  155. Song, S.; Ajani, J.A.; Honjo, S.; Maru, D.M.; Chen, Q.; Scott, A.W.; Heallen, T.R.; Xiao, L.; Hofstetter, W.L.; Weston, B.; et al. Hippo coactivator YAP1 upregulates SOX9 and endows esophageal cancer cells with stem-like properties. Cancer Res. 2014, 74, 4170–4182. [Google Scholar] [CrossRef] [PubMed]
  156. Ooki, A.; Del Carmen Rodriguez Pena, M.; Marchionni, L.; Dinalankara, W.; Begum, A.; Hahn, N.M.; VandenBussche, C.J.; Rasheed, Z.A.; Mao, S.; Netto, G.J.; et al. YAP1 and COX2 Coordinately Regulate Urothelial Cancer Stem-like Cells. Cancer Res. 2018, 78, 168–181. [Google Scholar] [CrossRef] [PubMed]
  157. Basu-Roy, U.; Bayin, N.S.; Rattanakorn, K.; Han, E.; Placantonakis, D.G.; Mansukhani, A.; Basilico, C. Sox2 antagonizes the Hippo pathway to maintain stemness in cancer cells. Nat. Commun. 2015, 6, 6411. [Google Scholar] [CrossRef]
  158. Zhao, X.; Sun, J.; Su, W.; Shan, H.; Zhang, B.; Wang, Y.; Shabanova, A.; Shan, H.; Liang, H. Melatonin Protects against Lung Fibrosis by Regulating the Hippo/YAP Pathway. Int. J. Mol. Sci. 2018, 19. [Google Scholar] [CrossRef]
  159. Perumal, N.; Perumal, M.; Halagowder, D.; Sivasithamparam, N. Morin attenuates diethylnitrosamine-induced rat liver fibrosis and hepatic stellate cell activation by co-ordinated regulation of Hippo/Yap and TGF-beta1/Smad signaling. Biochimie 2017, 140, 10–19. [Google Scholar] [CrossRef]
  160. Zhang, K.; Chang, Y.; Shi, Z.; Han, X.; Han, Y.; Yao, Q.; Hu, Z.; Cui, H.; Zheng, L.; Han, T.; et al. omega-3 PUFAs ameliorate liver fibrosis and inhibit hepatic stellate cells proliferation and activation by promoting YAP/TAZ degradation. Sci. Rep. 2016, 6, 30029. [Google Scholar] [CrossRef] [PubMed]
  161. Zhou, Y.; Huang, X.; Hecker, L.; Kurundkar, D.; Kurundkar, A.; Liu, H.; Jin, T.H.; Desai, L.; Bernard, K.; Thannickal, V.J. Inhibition of mechanosensitive signaling in myofibroblasts ameliorates experimental pulmonary fibrosis. J. Clin. Investig. 2013, 123, 1096–1108. [Google Scholar] [CrossRef] [PubMed]
  162. Bao, Y.; Nakagawa, K.; Yang, Z.; Ikeda, M.; Withanage, K.; Ishigami-Yuasa, M.; Okuno, Y.; Hata, S.; Nishina, H.; Hata, Y. A cell-based assay to screen stimulators of the Hippo pathway reveals the inhibitory effect of dobutamine on the YAP-dependent gene transcription. J. Biochem. 2011, 150, 199–208. [Google Scholar] [CrossRef] [PubMed]
  163. Sorrentino, G.; Ruggeri, N.; Specchia, V.; Cordenonsi, M.; Mano, M.; Dupont, S.; Manfrin, A.; Ingallina, E.; Sommaggio, R.; Piazza, S.; et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat. Cell Biol. 2014, 16, 357–366. [Google Scholar] [CrossRef] [PubMed]
  164. Ding, N.; Hah, N.; Yu, R.T.; Sherman, M.H.; Benner, C.; Leblanc, M.; He, M.; Liddle, C.; Downes, M.; Evans, R.M. BRD4 is a novel therapeutic target for liver fibrosis. Proc. Natl. Acad. Sci. USA 2015, 112, 15713–15718. [Google Scholar] [CrossRef] [PubMed]
  165. Yu, H.X.; Yao, Y.; Bu, F.T.; Chen, Y.; Wu, Y.T.; Yang, Y.; Chen, X.; Zhu, Y.; Wang, Q.; Pan, X.Y.; et al. Blockade of YAP alleviates hepatic fibrosis through accelerating apoptosis and reversion of activated hepatic stellate cells. Mol. Immunol. 2019, 107, 29–40. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The role of the Hippo pathway in the pathogenesis of tissue fibrosis. In normal cells, YAP/TAZ are phosphorylated and localized in the cytoplasm, leading to their inactivation. However, in many pathological situations, YAP/TAZ localize to the nucleus, and up-regulate their target genes to remodel the ECM. This action leads to tissue fibrosis. (a) In respiratory epithelial cells, binding YAP/TAZ with TTF-1 synergistically activates the expression of target genes, including SP-C, CTGF, and PAI-1. Also, active fibroblasts promote matrix synthesis via YAP/TAZ that are critical to the induction of profibrotic genes, such as CTGF and PAI-1, in the development of lung fibrosis. (b) NPHP4, by interacting with LATS1/2, promotes YAP/TAZ transcriptional activity. In contrast, NPHP9, another NPH proteins (NPHPs), directly interacts with YAP/TAZ and induces its nuclear localization to regulate a subset of target genes involved in renal fibrosis. (c) YAP/TAZ-mediated Indian hedgehog (Ihh) gene induction, which increases transcriptional reprogramming by modulating profibrotic gene expression in HSCs. In response to Ihh and TGF-β, HSCs transdifferentiate into the ECM-producing myofibroblasts. (d) YAP/TAZ are mainly localized with the nucleus, resulting in the profibrotic gene expression in dermal fibrolasts. See text for further details. The red spheres indicate the phosphorylation of YAP/TAZ by kinase.
Figure 1. The role of the Hippo pathway in the pathogenesis of tissue fibrosis. In normal cells, YAP/TAZ are phosphorylated and localized in the cytoplasm, leading to their inactivation. However, in many pathological situations, YAP/TAZ localize to the nucleus, and up-regulate their target genes to remodel the ECM. This action leads to tissue fibrosis. (a) In respiratory epithelial cells, binding YAP/TAZ with TTF-1 synergistically activates the expression of target genes, including SP-C, CTGF, and PAI-1. Also, active fibroblasts promote matrix synthesis via YAP/TAZ that are critical to the induction of profibrotic genes, such as CTGF and PAI-1, in the development of lung fibrosis. (b) NPHP4, by interacting with LATS1/2, promotes YAP/TAZ transcriptional activity. In contrast, NPHP9, another NPH proteins (NPHPs), directly interacts with YAP/TAZ and induces its nuclear localization to regulate a subset of target genes involved in renal fibrosis. (c) YAP/TAZ-mediated Indian hedgehog (Ihh) gene induction, which increases transcriptional reprogramming by modulating profibrotic gene expression in HSCs. In response to Ihh and TGF-β, HSCs transdifferentiate into the ECM-producing myofibroblasts. (d) YAP/TAZ are mainly localized with the nucleus, resulting in the profibrotic gene expression in dermal fibrolasts. See text for further details. The red spheres indicate the phosphorylation of YAP/TAZ by kinase.
Cells 08 00468 g001
Figure 2. The Hippo pathway drives fibrosis across the organs.
Figure 2. The Hippo pathway drives fibrosis across the organs.
Cells 08 00468 g002
Figure 3. Schematic model of regulation of the Hippo pathway by numerous signaling pathways in cancer. (a) Integrin-ILK signaling inhibits MYPT1, leading to inactivation of the MST1/2-LATS1/2 kinase cascade, which triggers YAP/TAZ activation. (b) RAC1 and PAK1 phosphorylate NF2 that inhibits the interaction between YAP and NF2, resulting in YAP/TAZ activation. (c) Integrin-FAK signaling activates the RhoA-ROCK pathway to control remodeling of the actin cytoskeleton. (d) In response to ECM, Agrin enhances RhoA-ROCK-dependent actin cytoskeletal remodeling. As a later event, YAP/TAZ are translocated to the nucleus to interact with transcription factors (TFs), which drive profibrotic and tumorigenic effects.
Figure 3. Schematic model of regulation of the Hippo pathway by numerous signaling pathways in cancer. (a) Integrin-ILK signaling inhibits MYPT1, leading to inactivation of the MST1/2-LATS1/2 kinase cascade, which triggers YAP/TAZ activation. (b) RAC1 and PAK1 phosphorylate NF2 that inhibits the interaction between YAP and NF2, resulting in YAP/TAZ activation. (c) Integrin-FAK signaling activates the RhoA-ROCK pathway to control remodeling of the actin cytoskeleton. (d) In response to ECM, Agrin enhances RhoA-ROCK-dependent actin cytoskeletal remodeling. As a later event, YAP/TAZ are translocated to the nucleus to interact with transcription factors (TFs), which drive profibrotic and tumorigenic effects.
Cells 08 00468 g003
Figure 4. YAP/TAZ-mediated regulatory networks in cancer. YAP/TAZ are the major downstream mediators of the Hippo pathway and are composed of several domains (see Figure 1). The nuclear YAP/TAZ interacts with multiple transcription factors and signaling pathway that collectively contribute to tumorigenesis.
Figure 4. YAP/TAZ-mediated regulatory networks in cancer. YAP/TAZ are the major downstream mediators of the Hippo pathway and are composed of several domains (see Figure 1). The nuclear YAP/TAZ interacts with multiple transcription factors and signaling pathway that collectively contribute to tumorigenesis.
Cells 08 00468 g004
Table 1. Potential targets for effective Hippo pathway regulation.
Table 1. Potential targets for effective Hippo pathway regulation.
NameMode of ActionTested ApplicationReference
VerteporfinDisruption of YAP/TAZ- TEAD complex
  • HSCs isolated from C57BL/6J mice or Smo flox/flox mice
  • IPF lung epithelial cells
  • UUO-induced renal fibrosis
  • CCl4-induced liver fibrosis mouse model
[49,63,68]
MelatoninInhibiting of the expression and activation of YAP1 via binding to MT1&MT2 melatonin receptors
  • Bleomycin induced experimental lung fibrosis in mice
[158]
MorinIncreased expression of MST1 and Lats1 Decreased expression of YAP/TAZ
  • Diethylnitrosamine (Den)-induced liver fibrosis rat model
  • Hepatic stellate cells derived form human
[159]
ω-3 PUFAYAP/TAZ degradation in a
proteasome-dependent manner
  • CCl4-induced liver fibrosis mouse model
  • Hepatic stellate cells derived from human and rat
[160]
FasudilInhibitory effect on Rho/ROCK signaling
  • Bleomycin-induced mouse lung fibrosis
[161]
DobutamineInduced the cytoplasmic translocation of YAP
  • U2OS cell line
[162]
StatinYAP/TAZ nuclear localization and activity
  • Breast cancer cell lines
[163]
LovastatinYAP/TAZ nuclear localization and activity
  • CCl4-induced liver fibrosis mouse model
  • Human aortic vascular smooth muscle cells
  • Human kidney cells
[79]
JQ-1Inhibitor of bromodomain-containing protein 4 (BRD4) mediated profibrotic transcription
  • CCl4-induced liver fibrosis mouse model
  • Hepatic stellate cells derived from mouse
[164]

Share and Cite

MDPI and ACS Style

Kim, C.-L.; Choi, S.-H.; Mo, J.-S. Role of the Hippo Pathway in Fibrosis and Cancer. Cells 2019, 8, 468. https://0-doi-org.brum.beds.ac.uk/10.3390/cells8050468

AMA Style

Kim C-L, Choi S-H, Mo J-S. Role of the Hippo Pathway in Fibrosis and Cancer. Cells. 2019; 8(5):468. https://0-doi-org.brum.beds.ac.uk/10.3390/cells8050468

Chicago/Turabian Style

Kim, Cho-Long, Sue-Hee Choi, and Jung-Soon Mo. 2019. "Role of the Hippo Pathway in Fibrosis and Cancer" Cells 8, no. 5: 468. https://0-doi-org.brum.beds.ac.uk/10.3390/cells8050468

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

Article Metrics

Back to TopTop