Hippo Cancer

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Review article 1

Two faces of Hippo: activate or suppress the Hippo pathway


in cancer
Jingwen Cao and Wenlong Huang

The Hippo pathway has generated considerable interest in strategy for cancer treatment. This review will provide a
recent years because of its involvement in several key summary of currently known compounds that activate or
hallmarks of cancer progression and metastasis. Research suppress the Hippo pathway. Anti-Cancer Drugs
on the Hippo signaling pathway in cancer has been used to 00:000–000 Copyright c 2017 Wolters Kluwer Health, Inc.
determine the activity of yes-associated protein (YAP) in All rights reserved.
tumorigenesis and disease progression. Previous studies Anti-Cancer Drugs 2017, 00:000–000
have shown that the Hippo pathway can be used as a target
to inhibit YAP activity and is a viable treatment for cancer. Keywords: Hippo pathway, tumor immune response, tumorigenesis

However, more studies are required to further advance our School of International Pharmaceutical Business, China Pharmaceutical
understanding of the Hippo signaling pathway in cancer. It University, Nanjing, China

has been shown that knockout of serine/threonine-kinases Correspondence to Wenlong Huang, PhD, China Pharmaceutical University,
LATS1/2 in the Hippo pathway suppresses cancer immunity #24 TongJiaXiang, Gulou District, Nanjing 210009, China
Tel: + 86 258 327 1487; fax: + 86 2583 271 512;
in mice. In addition, suppression of the oncogene YAP could e-mail: [email protected]
contribute toward cancer immune therapy. Therefore,
Received 26 June 2017 Revised form accepted 17 August 2017
regulation of Hippo signaling can be an attractive alternative

Introduction Central to the Hippo pathway is a protein kinase cascade.


Hippo signaling is an evolutionarily conserved pathway Once upstream activation signals are received, a complex
that controls organ size by regulating cell proliferation, formed by MST1/2 kinases and SAV1 phosphorylates and
apoptosis, and stem cell self-renewal [1]. Dysregulation activates LATS1/2, which in turn phosphorylate the tran-
of this signaling pathway contributes toward cancer scription co-activators YAP/TAZ [7]. Phosphorylation indu-
development [2]. In this review, we will provide a brief ces 14-3-3 binding and cytoplasmic retention of YAP [7].
introduction to the Hippo signaling pathway, with a focus This leads to spatial separation between YAP/TAZ and their
on components that are altered in human cancers. We nuclear target transcription factors [8]. In addition, YAP
will then focus on potential cancer therapies based on the phosphorylation also leads to β-transducin repeat-containing
Hippo pathway, and present issues and future directions E3 ubiquitin protein ligase-dependent degradation by pro-
for Hippo pathway-based anticancer strategies. teasomes [9,10]. However, once dephosphorylated, YAP/
TAZ translocate to the nucleus and interact with TEAD1-4,
as well as other transcription factors, to promote the
Components of the Hippo pathway expression of genes that contribute toward cell proliferation
The Hippo signaling pathway, also known as the and survival [11] (Fig. 1).
Salvador/Warts/Hippo pathway, has emerged recently as
a growth control pathway. It was first discovered in
Drosophila melanogaster, and was later shown to be highly Role of the Hippo pathway in cancer
conserved in humans [3,4]. The Hippo pathway consists Deregulation of the Hippo pathway in cancer
of a core serine–threonine kinase cascade and a tran- There is considerable evidence to suggest that the Hippo
scriptional module. The kinase cascade includes mam- pathway is frequently deregulated in a broad range of
malian STE-20-like protein kinases 1/2 (MST1/2) and human cancers, including breast, ovarian, colorectal, lung,
large tumor-suppressor kinases 1/2 (LATS1/2) [5]. The and liver cancers; this is often correlated with poor
transcriptional co-factors, yes-associated protein (YAP), patient prognosis [12–14].
and transcriptional co-activator with PDZ-binding motif
(TAZ) together form the transcriptional module [6]. YAP primarily resides in the cytoplasm in normal human
Upon entering the nucleus, YAP/TAZ bind to the tran- tissues. Immunohistochemical staining of tumor tissues
scription factor TEAD and upregulate genes important indicated that deregulation of the Hippo pathway results
for proliferation and cell survival. It has been shown that in prolonged nuclear localization of YAP. Binding of YAP
YAP and TAZ are the main proteins that modulate sig- to TEAD initiates growth and survival-associated tran-
naling in the Hippo pathway [6]. scriptional programs.
0959-4973 Copyright
c 2017 Wolters Kluwer Health, Inc. All rights reserved. DOI: 10.1097/CAD.0000000000000559

Copyright r 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
2 Anti-Cancer Drugs 2017, Vol 00 No 00

Fig. 1 ubiquitin ligase CRL4 (DCAF1). This consequently


inhibits ubiquitination of LATS1/2 and inactivates YAP.
It is still unclear why mutations in Hippo signaling
pathway genes are so rare as YAP and TAZ activation is
observed in a broad range of human cancers. It is possible
that cross-networks such as Wnt [34], transforming
growth factor-β–bone morphogenetic protein (TGF-
β–BMP) [35], Notch [36], epithelial growth factor
receptor (EGFR) [37], and G-protein-coupled receptor
(GPCR) [38] frequently harbor oncogenic mutations,
which affect YAP/TAZ activation. Activation of these
pathways has been correlated with immunohistochemical
detection of nuclear YAP and TAZ in tumor tissues
[39–43]. Further investigation is required to define the
major oncogenic sources that cause YAP and TAZ acti-
vation in various tumor types.
Epigenetic alteration may be another mechanism by
which YAP and TAZ are activated in some human
tumors. Approximately 50% of breast cancers and
60–70% of astrocytomas have been shown to have
hypermethylated LATS1/2 promoter regions that limit
the expression of these genes [23,24]. In 37% of soft
tissue sarcomas and 20% of sarcomas, the MST1/2 pro-
Core of the Hippo pathway. moter regions were also shown to be hypermethylated
[44]. In addition, YAP amplification was observed on
chromosome 11q22 amplicons [16]. These results sug-
gested that downregulation of Hippo signaling by epi-
Elevated levels and nuclear localization of YAP have
genetic silencing of key activators and amplification of
been reported in a majority of solid cancers; these factors
YAP is associated with various lethal cancers.
are also strongly associated with adverse overall survival
and disease-free survival time in numerous cancers,
Hippo pathway as a suppressor signal for
suggesting that YAP1 may act as a potential therapeutic
tumorigenesis
target for these malignancies [12,14–20].
YAP/TAZ are pervasively activated in many human
tumors [12,45,46]. Elevated YAP and TAZ activity was
Potential mechanisms of Hippo pathway deregulation shown to induce tumors in several mouse models. In a
in cancer YAP transgenic model, overexpression of YAP con-
Approximately 50% of human liver cancers result in tributes toward the development of hepatocellular carci-
elevated YAP levels [21] and TAZ overexpression is noma [47], promotes irregular expansion in liver tissues,
observed in over 80% of breast cancers [19,22]. These and is associated with undifferentiated intestinal pro-
findings suggest that amplification of YAP and TAZ is a genitor cells [15]. Furthermore, YAP overexpression in
common mechanism underlying some human cancers. the KRASG12D lung cancer mouse model accelerated lung
However, alternative mechanisms predominate in other adenocarcinomas progression [48]. YAP overexpression
types of cancers, and could include mutation of Hippo also contributes toward radiation resistance by induction
pathway genes, promoter methylation, epigenetic silen- of genomic instability [49]. Therefore, YAP activation
cing of MST and LATS kinases [23–25], and expression confers potent cell proliferative capacity and resistance to
and/or upregulation of proteins that control Yap tran- apoptosis, which can eventually lead to cancer develop-
scription [26–29] and stability [28]. ment [46]. In addition, TAZ-induced transcriptional
activation of ZEB1/2 stimulates epithelial-to-mesenchy-
Recent reports on the basis of data from The Cancer
mal transition [50,51].
Genome Atlas and Catalogue of Somatic Mutation in
Cancer census suggested that Hippo pathway genes, with Mouse models with deletions in MST1/2, SAV1, MOB1A/
the exception of NF2 and TAZ, are not cancer-related B, and LATS1/2 all show upregulated expression of
genes [12]. NF2 encodes the tumor-suppressor protein TEAD target genes, increased expansion of progenitor
merlin, which regulates the Hippo pathway by promoting cells, as well as tissue overgrowth or tumorigenesis.
localization of LATS kinases to the plasma membrane Combined MST1/2 deficiency in the liver results in loss
[30–32]. Li et al. [33] found that merlin could bind to of inhibitory Ser127 phosphorylation on YAP1, massive
DCAF1 in the nucleus to suppress the activity of the E3 cell overgrowth, and hepatocellular carcinoma [52–54].

Copyright r 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
Two faces of Hippo Cao and Huang 3

In addition, studies have shown that in liver-specific YAP and TAZ through TEAD-mediated transcription is
LATS1/2-knockout and MOB1-deficient mice, the liver is required for in-vivo tumor growth suppression by
filled with highly proliferated immature biliary epithelial LATS1/2 deletion. This study uncovered a key role of the
cells [39,55]. Moreover, the deletion of SAV1 in hepato- Hippo pathway in modulating tumor immunogenicity,
cytes and intestinal epithelial cells leads to an increase in and showed that activation of the Hippo pathway may be
hepatic and intestinal progenitor cells, respectively [56,57]. a potential form of tumor therapy.
In the hypomorphic MOB1 mouse model, various types of
Activating the Hippo pathway may promote antitumor
tumors are developed [58]. Dysregulation of the Hippo
immune response, and therefore, would be an attractive
signaling pathway and increased YAP/TAZ activity induce
approach for cancer treatment. Furthermore, LATS1/2
expansion of tissue-specific stem/progenitor cells that
inhibition may enhance the efficacy of immune check-
eventually leads to cancer development. Therefore, clar-
point inhibitors. Thus, a combination of Hippo pathway
ification of the potential roles of the Hippo signaling
activators and immune checkpoint inhibitors could be a
pathway in tissue homeostasis and cancer stem cells can aid
novel therapeutic approach for immunogenic cancers,
in the advancement of cancer therapy.
especially in cases where malignancy is driven by onco-
genic alterations that do not affect the Hippo signaling
Hippo pathways have different tissue-specific and cell-
pathway. As germline or somatic mutations that affect the
type-specific physiological roles
core components of the Hippo pathway are uncommon in
It is generally accepted that the Hippo pathway is a
human cancers, it is expected that activation of the Hippo
tumor suppressor that inhibits the proliferation and sur-
pathway can enhance tumor immunity in most cancer
vival of normal cells, thus preventing tumorigenesis
types (Fig. 2).
[10,12,59]. However, a few studies have suggested that
the Hippo pathway can also exert oncogenic effects
under certain contexts [60,61]. Therapeutic targets in the Hippo pathway: reactivation of
the Hippo pathway
The correlation between Hippo signaling and cancer is Targeting kinases
tissue and cell specific. For example, the epidermis of Generally, kinases are the best targets for small molecule
mice with a skin-specific deletion of MST1/2 or LATS1/2 therapeutics. Unlike most of the oncogenic kinase targets
showed normal morphology, and YAP-S127 phosphor- used in current cancer therapies, the majority of kinases
ylation was not increased [62]. In addition, no kidney in the Hippo pathway are tumor suppressors. Restoring
defects or YAP activation were observed following the the function of tumor-suppressor kinases is indeed a
deletion of MST1/2 or SAV1 [63]. These studies indicated challenging task; activation of MST or LATS kinases for
that YAP activity is not always elevated during restriction anticancer therapy is even more challenging as there are
of core Hippo components. Similarly, it has been shown few options for the rational design of small molecule
that the oncogenic phenotype does not always occur with agonists [65].
YAP activation. It was observed that intestinal epithelial
cell-specific YAP expression leads to rapid loss of Targeting F-actin
intestinal crypts and inhibition of Wnt-mediated intest- F-actin modulates YAP/TAZ nuclear translocation,
inal regeneration [60]. These results suggested that the allowing cells to upregulate proliferation and survival
Hippo signaling pathway and YAP/TAZ exert different genes [66–68]. Therefore, it is reasonable to speculate
tissue-specific or cell-type-specific effects. that drugs that target F-actin may be effective in control
of tumor growth. The F-actin destabilizers, cytochalasin
The Hippo pathway kinases LATS1/2 suppress cancer D, the RHO kinase inhibitor Y27632, and the nonmuscle
immunity myosin II inhibitor blebbistatin all promote nuclear
For decades, most research groups have focused on the export of YAP and TAZ [69–72]. However, the impor-
suppressor role of the Hippo pathway in cancer. tance of the actin cytoskeleton for various basic cellular
However, the role of the Hippo pathway in the context of functions places limits on their usage. Therefore, appro-
reciprocal interactions between tumor cells and host priate doses of F-actin inhibitors need to be chosen to not
antitumor immune responses remains largely unknown. only show therapeutic efficacy but also to produce
Moroishi et al. [64] reported that loss of the Hippo minimal side effects.
pathway kinases LATS1/2 promotes tumor cell growth
in vitro, but inhibits tumor growth in murine syngeneic Targeting the GPCR signaling pathway
tumor models. Furthermore, adaptive immune responses GPCRs are the largest and most diverse group of mem-
induced by LATS1/2 deletion cause tumor regression. brane receptors in eukaryotes. These cell surface recep-
Mechanistically, it has been shown that nucleic acid-rich tors function like an inbox for messages in the form of
extracellular vesicles secreted by LATS1/2-null tumor light energy, peptides, lipids, sugars, and proteins.
cells induce type I interferon responses by the Toll-like Yu et al. [73] reported that YAP/TAZ is regulated robustly
receptor-MYD88/TRIF pathway. Hyperactivation of by many GPCRs and their cognate ligands, and

Copyright r 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
4 Anti-Cancer Drugs 2017, Vol 00 No 00

Fig. 2

Role of the Hippo pathway in cancer.

established the general function of GPCRs in YAP/TAZ enhancer element within the first intron of the YAP gene
regulation. GPCRs usually activate downstream signaling to drive YAP expression. In addition, detachment of β-
through heterotrimeric G proteins. Gα12/13-coupled, catenin from the destruction complex formed by APC,
Gαq/11-coupled, and Gαi/o-coupled signals induce YAP/ AXIN, and GSK3 resulted in inhibition of YAP/TAZ
TAZ activity, whereas Gαs-coupled signals repress YAP/ degradation [74]. Therefore, targeting the Wnt pathway
TAZ activity. Stimulation of Gs-coupled receptors by would be an effective option for inhibition of YAP/TAZ
glucagon or epinephrine activates LATS1/2 kinase transcription. Tankyrase inhibitors such as G244-LM,
activity, thereby inhibiting YAP function. Therefore, G007-LK, and XAV939 could increase AXIN activity and
development of small-molecule agonists targeting Gαs restore the integrity of the β-catenin destruction com-
may be an effective therapeutic option. plex, leading to YAP/TAZ degradation [75–77].

Targeting the Wnt/β-catenin signaling pathway YAP and TEAD as therapeutic targets
Konsavage et al. [29] implicated YAP as an oncogene Functionally, the most attractive therapeutic targets in
whose expression is driven by aberrant Wnt/β-catenin the Hippo pathway are the key oncoproteins, YAP and
signaling in human CRC cells. Once Wnt signaling is TAZ. YAP and TAZ are the final common conduits of
turned on, β-catenin/TCF4 complexes bind to a DNA the Hippo signaling pathway, and transmit signals from

Copyright r 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
Two faces of Hippo Cao and Huang 5

the core kinase cassette and upstream regulatory proteins upregulate YAP and TAZ function, which may prove
to a progrowth transcriptional program. beneficial in activation of the Hippo pathway.

Verteporfin Targeting MST1


Liu-Chittenden et al. [78] screened a small-molecule Anand et al. [84] reported the development of a potent
FDA-approved drug library for targeting the integrity of and selective MST1 kinase inhibitor based on a ruthe-
the TEAD–YAP transcription factor complex. Three nium half-sandwich scaffold. This compound was shown
compounds (protoporphyrin IX, hematoporphyrin, and to inhibit MST activity in vitro and in cultured Hela cells,
verteporfin) stood out among the top hits, all of which as measured by Histone H2B phosphorylation in
belong to the porphyrin family. Verteporfin, a benzo- response to apoptosis. However, its effects on YAP/TAZ
porphyrin derivative, is in clinical use as a photosensitizer activity have not yet been reported. The 9E1 antibody
in photocoagulation therapy for macular degeneration shows significant, although not complete, selectivity for
[79]. More importantly, verteporfin was moderately MST1 over other kinases, and provides a starting point
effective in blocking mouse hepatic tumorigenesis driven for the development of more selective MST1 inhibitors.
by Yap1 overexpression or loss of Nf2 [78], suggesting MST1 and MST2 (MST1/2) kinases, the mammalian
that it functions downstream of both of these alterations. Hippo orthologs, are central components of the Hippo
This strategy may be particularly appealing for tumors signaling pathway, and are therefore ideal target candi-
that are linked to mutations in Nf2, such as brain and dates for pharmacological induction of the Hippo pathway
neural tumors. [85]. Fan and colleagues reported the discovery of a
reversible and selective MST1/2 inhibitor, XMU-MP-1,
VGLL4-mimicking peptide using an enzyme-linked immunosorbent assay-based high-
VGLL4 directly competes with YAP in binding to throughput biochemical assay. XMU-MP-1 showed
TEADs, and serves its growth-inhibitory function excellent in-vivo pharmacokinetics, and could promote
through two TDU domains [80,81]. On the basis of the mouse intestinal repair. In addition, an intraperitoneal
structure of the VGLL4–TEAD4 complex, Jiao et al. [82] injection of XMU-MP-1 stimulated liver repair and
developed a peptide-based YAP inhibitor that mimics the regeneration in both acute and chronic liver injury mouse
function of VGLL4. This peptide could suppress tumor models at a dose of 1–3 mg/kg. The next step is to utilize
growth of human primary gastric cancer in nude mice and XMU-MP-1 for tumor therapies. We predict that future
inhibit tumorigenesis in the Helicobacter pylori-infected tumor therapies that use Hippo pathway activators will be
mouse model of gastric cancer [82]. heavily dependent on immune responses.

Flufenamic acid Targeting GPCR


Pobbati et al. [83] reported the discovery of a central GPCRs usually activate downstream signaling through
pocket in the YAP-binding domain (YBD) of TEAD. heterotrimeric G proteins. Yu et al. [73] showed that sti-
Screening of compounds that target this central pocket in mulation of Gα12/13-coupled, Gαq/11-coupled, and Gαi/
YBD was performed by differential scanning fluorimetry. o-coupled signals with lysophosphatidic acid (LPA),
X-ray crystallography studies showed that flufenamic sphingosine 1-phosphophate, and thrombin induced
acid, a NSAID, binds to the central pocket of TEAD2 YAP/TAZ activity. Serum-borne LPA and sphingosine
YBD. Biochemical and functional analyses further 1-phosphophate act through G12/13-coupled receptors to
showed that binding of NSAIDs to TEAD inhibits inhibit the Hippo pathway kinases, LATS1/2. This
TEAD–YAP-dependent transcription, cell migration, and results in the activation of the transcription co-activators,
proliferation, indicating that the central pocket is YAP and TAZ, which are oncoproteins repressed by
important for TEAD function. These studies pointed to a LATS1/2. YAP and TAZ are involved in LPA-induced
novel approach to targeting TEAD transcription factors, gene expression, cell migration, and proliferation [86].
and set the stage for therapeutic development of specific
TEAD–YAP inhibitors against human cancers. Discussion
High YAP and TAZ activity has been shown in various
Therapeutic targets in the Hippo pathway: suppression types of cancer. However, genetic mutations in the
of the Hippo pathway Hippo core components, YAP and TAZ, are rare [87].
Targeting kinase This implies that aberrant activation of the Hippo path-
Decades of targeted drug development efforts suggest way may be insufficient for tumorigenesis in vivo.
that kinases and enzymes are the most attractive targets
for small-molecule therapies. MST1/2 and LATS1/2 are Combination therapy
two pairs of kinases at the core of the Hippo signaling Activation of YAP may be triggered by other biological
pathway, and act to restrain YAP and TAZ activity. Small- processes that are sensitive to oncogenic mutations.
molecule inhibitors of these kinases are predicted to Understanding the synergistic effects between the Hippo

Copyright r 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
6 Anti-Cancer Drugs 2017, Vol 00 No 00

pathway and processes that result in tumorigenesis could 17 Zender L, Spector MS, Xue W, Flemming P, Cardon-Cardo C, Silke J, et al.
advance our knowledge of the Hippo pathway. Identification and validation of oncogenes in liver cancer using an integrative
oncogenomic approach. Cell 2006; 125:1253–1267.
Combination therapies that target both Hippo signaling 18 Steinhardt AA, Gayyed MF, Klein AP, Dong J, Maitra A, Pan D, et al.
and these auxiliary pathways could be an attractive Expression of Yes-associated protein in common solid tumors. Hum Pathol
approach for cancer therapy. 2008; 39:1582–1589.
19 Chan SW, Lim CJ, Guo K, Ng CP, Lee I, Hunziker W, et al. A role for TAZ in
migration, invasion, and tumorigenesis of breast cancer cells. Cancer Res
2008; 68:2592–2598.
Targeting the Hippo pathway for cancer immunotherapy 20 Zhao B, Wei X, Li W, Udan RS, Yang Q, Kim J, et al. Inactivation of YAP
Recent advancements in cancer immunotherapy have set oncoprotein by the Hippo pathway is involved in cell contact inhibition and
the stage for novel therapeutic strategies; several immune tissue growth control. Genes Dev 2007; 21:2747–2761.
21 Xu MZ, Yao TJ, Lee NP, Ng IO, Chan Y-T, Zender L, et al. Yes-associated
checkpoint modulators such as PD-1 inhibitor have protein is an independent prognostic marker in hepatocellular carcinoma.
shown impressive effects in clinic trials [88]. However, Cancer 2009; 115:4576–4585.
immune checkpoint inhibitors may not work in cases 22 Cordenonsi M, Zanconato F, Azzolin L, Forcato M, Rosato A, Frasson C,
et al. The Hippo transducer TAZ confers cancer stem cell-related traits on
where tumor immunogenicity is intrinsically generated breast cancer cells. Cell 2011; 147:759–772.
[89]. Therefore, a combination of immune checkpoint 23 Jiang Z, Li X, Hu J, Zhou W, Jiang Y, Li G, Lu D. Promoter hypermethylation-
inhibitors and Hippo pathway suppressors could be an mediated down-regulation of LATS1 and LATS2 in human astrocytoma.
Neurosci Res 2006; 56:450–458.
exciting therapeutic approach, especially for poorly con- 24 Takahashi Y, Miyoshi Y, Takahata C, Irahara N, Taguchi T, Tamaki Y,
trolled immunogenic cancers driven by other oncogenic Noguchi S. Down-regulation of LATS1 and LATS2 mRNA expression by
alterations that do not affect the Hippo signaling path- promoter hypermethylation and its association with biologically aggressive
phenotype in human breast cancers. Clin Cancer Res 2005;
way [64]. 11:1380–1385.
25 Li H, Wolfe A, Septer S, Edwards G, Zhong X, Abdulkarim AB, et al.
Deregulation of Hippo kinase signalling in human hepatic malignancies. Liver
Acknowledgements Int 2012; 32:38–47.
26 Zhang T, Zhang J, You X, Liu Q, Du Y, Gao Y, et al. Hepatitis B virus X protein
Conflicts of interest
modulates oncogene Yes-associated protein by CREB to promote growth of
There are no conflicts of interest. hepatoma cells. Hepatology 2012; 56:2051–2059.
27 Wu H, Xiao Y, Zhang S, Ji S, Wei L, Fan F, et al. The Ets transcription factor
GABP is a component of the hippo pathway essential for growth and
antioxidant defense. Cell Rep 2013; 3:1663–1677.
Reference 28 Wang J, Park JS, Wei Y, Rajurkar M, Cotton JL, Fan Q, et al. TRIB2 acts
1 Zhao B, Tumaneng K, Guan KL. The Hippo pathway in organ size control,
downstream of Wnt/TCF in liver cancer cells to regulate YAP and C/
tissue regeneration and stem cell self-renewal. Nat Cell Biol 2011;
EBPalpha function. Mol Cell 2013; 51:211–225.
13:877–883.
29 Konsavage WM Jr, Kyler SL, Rennoll SA, Jin G, Yochum GS. Wnt/beta-
2 Pan D. The hippo signaling pathway in development and cancer. Dev Cell
catenin signaling regulates Yes-associated protein (YAP) gene expression in
2010; 19:491–505.
colorectal carcinoma cells. J Biol Chem 2012; 287:11730–11739.
3 Badouel C, McNeill H. SnapShot: The hippo signaling pathway. Cell 2011;
30 Hamaratoglu F, Willecke M, Kango-Singh M, Nolo R, Hyun E, Tao C, et al.
145:484.e1–484.e1.
The tumour-suppressor genes NF2/Merlin and Expanded act through Hippo
4 Meignin C, Alvarez-Garcia I, Davis I, Palacios IM. The salvador-warts-hippo
signalling to regulate cell proliferation and apoptosis. Nat Cell Biol 2006;
pathway is required for epithelial proliferation and axis specification in
8:27–36.
Drosophila. Curr Biol 2007; 17:1871–1878. 31 Zhang N, Bai H, David KK, Dong J, Zheng Y, Cai J, et al. The Merlin/NF2
5 Mo JS, Park HW, Guan KL. The Hippo signaling pathway in stem cell biology tumor suppressor functions through the YAP oncoprotein to regulate tissue
and cancer. EMBO Rep 2014; 15:642–656. homeostasis in mammals. Dev Cell 2010; 19:27–38.
6 Zanconato F, Cordenonsi M, Piccolo S. YAP/TAZ at the roots of cancer. 32 Yin F, Yu J, Zheng Y, Chen Q, Zhang N, Pan D. Spatial organization of Hippo
Cancer Cell 2016; 29:783–803. signaling at the plasma membrane mediated by the tumor suppressor Merlin/
7 Meng Z, Moroishi T, Guan KL. Mechanisms of Hippo pathway regulation. NF2. Cell 2013; 154:1342–1355.
Genes Dev 2016; 30:1–17. 33 Li W, Cooper J, Zhou L, yang C, Erdjument-Bromage H, Zagzag D, et al.
8 Zhao B, Li L, Lei Q, Quan KL. The Hippo-YAP pathway in organ size control Merlin/NF2 loss-driven tumorigenesis linked to CRL4(DCAF1)-mediated
and tumorigenesis: an updated version. Genes Dev 2010; 24:862–874. inhibition of the hippo pathway kinases Lats1 and 2 in the nucleus. Cancer
9 Johnson R, Halder G. The two faces of Hippo: targeting the Hippo pathway Cell 2014; 26:48–60.
for regenerative medicine and cancer treatment. Nat Rev Drug Discov 2014; 34 White BD, Chien AJ, Dawson DW. Dysregulation of Wnt/beta-catenin
13:63–79. signaling in gastrointestinal cancers. Gastroenterology 2012; 142:219–232.
10 Moroishi T, Park HW, Qin B, Chen Q, Meng Z, Plouffe SW, et al. A YAP/TAZ- 35 Bellam N, Pasche B. Tgf-beta signaling alterations and colon cancer. Cancer
induced feedback mechanism regulates Hippo pathway homeostasis. Genes Treat Res 2010; 155:85–103.
Dev 2015; 29:1271–1284. 36 Lobry C, Oh P, Aifantis I. Oncogenic and tumor suppressor functions of
11 Lamar JM, Stern P, Liu H, Schindler JW, et alJiang ZG, et alHynes RO. The Notch in cancer: it's NOTCH what you think. J Exp Med 2011;
Hippo pathway target, YAP, promotes metastasis through its TEAD- 208:1931–1935.
interaction domain. Proc Natl Acad Sci USA 2012; 109:E2441–E2450. 37 Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR
12 Harvey KF, Zhang X, Thomas DM. The Hippo pathway and human cancer. mutations in lung cancer activate anti-apoptotic pathways. Science 2004;
Nat Rev Cancer 2013; 13:246–257. 305:1163–1167.
13 Moroishi T, Hansen CG, Guan KL. The emerging roles of YAP and TAZ 38 Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L,
in cancer. Nat Rev Cancer 2015; 15:73–79. O’Brien JM, et al. Frequent somatic mutations of GNAQ in uveal melanoma
14 Sun Z, Xu R, Li X, Ren W, Ou C, Wang Q, et al. Prognostic value of yes- and blue naevi. Nature 2009; 457:599–602.
associated protein 1 (YAP1) in various cancers: a meta-analysis. PLoS One 39 Nishio M, Sugimachi K, Goto H, Wang J, Morikawa T, Miyachi Y, et al.
2015; 10:e0135119. Dysregulated YAP1/TAZ and TGF-beta signaling mediate
15 Camargo FD, Gokhale S, Johnnidis JB, Fu D, Bell GW, Jaenisch R, hepatocarcinogenesis in Mob1a/1b-deficient mice. Proc Natl Acad Sci USA
Brummelkamp TR, Wang Q. YAP1 increases organ size and expands 2016; 113:E71–E80.
undifferentiated progenitor cells. Curr Biol 2007; 17:2054–2060. 40 Tschaharganeh DF, Chen X, Latzko P, Malz M, Gaida MM, Felix K, et al. Yes-
16 Overholtzer M, Zhang J, Smolen GA, Muir B, Li W, Sgroi DC, et al. associated protein up-regulates Jagged-1 and activates the Notch pathway
Transforming properties of YAP, a candidate oncogene on the chromosome in human hepatocellular carcinoma. Gastroenterology 2013; 144:1530.
11q22 amplicon. Proc Natl Acad Sci USA 2006; 103:12405–12410. e12–1542.e12.

Copyright r 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
Two faces of Hippo Cao and Huang 7

41 Rosenbluh J, Nijhawan D, Cox AG, Li X, Neal JT, Schafer EJ, et al. β-Catenin- 66 Sansores-Garcia L, Bossuyt W, Wada K, Yonemura S, Tao C, Sasaki H,
driven cancers require a YAP1 transcriptional complex for survival and Halder G. Modulating F-actin organization induces organ growth by affecting
tumorigenesis. Cell 2012; 151:1457–1473. the Hippo pathway. EMBO J 2011; 30:2325–2335.
42 Yang N, Morrison CD, Liu P, J Miecznikowski, Bshara W, Han S, et al. TAZ 67 Reddy P, Deguchi M, Cheng Y, Hsueh AJ. Actin cytoskeleton regulates
induces growth factor-independent proliferation through activation of EGFR Hippo signaling. PLoS One 2013; 8:e73763.
ligand amphiregulin. Cell Cycle 2012; 11:2922–2930. 68 Mana-Capelli S, Paramasivam M, Dutta S, McCollum D. Angiomotins link
43 Zhou X, Wang S, Wang Z, Feng X, Liu P, Lv XB, et al. Estrogen regulates F-actin architecture to Hippo pathway signaling. Mol Biol Cell 2014;
Hippo signaling via GPER in breast cancer. J Clin Invest 2015; 25:1676–1685.
125:2123–2135. 69 Muehlich S, Rehm M, Ebenau A, Goppelt-Struebe M. Synergistic induction
44 Seidel C, Schagdarsurengin U, Blümke K, Würl P, Pfeifer GP, Hauptmann S, of CTGF by cytochalasin D and TGFbeta-1 in primary human renal epithelial
et al. Frequent hypermethylation of MST1 and MST2 in soft tissue sarcoma. cells: role of transcriptional regulators MKL1, YAP/TAZ and Smad2/3. Cell
Mol Carcinog 2007; 46:865–871. Signal 2017; 29:31–40.
45 Piccolo S, Dupont S, Cordenonsi M. The biology of YAP/TAZ: hippo 70 Sen B, Xie Z, Uzer G, Thompson WR, Styner M, Wu X, Rubin J. Intranuclear
signaling and beyond. Physiol Rev 2014; 94:1287–1312. actin regulates osteogenesis. Stem Cells 2015; 33:3065–3076.
46 Zanconato F, Battilana G, Cordenonsi M, Piccolo S. YAP/TAZ as therapeutic 71 Dupont S, Morsut L, Aragona M, Enzo E, Giulitti S, Cordenonsi M, et al. Role
targets in cancer. Curr Opin Pharmacol 2016; 29:26–33. of YAP/TAZ in mechanotransduction. Nature 2011; 474:179–183.
47 Bai H, Gayyed MF, Lam-Himlin DM, Klein AP, Nayar SK, Xu Y, et al.
72 Das A, Fischer RS, Pan D, Waterman CM. YAP nuclear localization in the
Expression of Yes-associated protein modulates Survivin expression in
absence of cell–cell contact is mediated by a filamentous actin-dependent,
primary liver malignancies. Hum Pathol 2012; 43:1376–1385.
myosin II- and phospho-YAP-independent pathway during extracellular matrix
48 Zhang W, Gao Y, Li F, Tong X, Ren Y, Han X, et al. YAP promotes malignant
mechanosensing. J Biol Chem 2016; 291:6096–6110.
progression of Lkb1-deficient lung adenocarcinoma through downstream
73 Yu FX, Zhao B, Panupinthu N, Jewell JL, Lian I, Wang LH, et al. Regulation of
regulation of survivin. Cancer Res 2015; 75:4450–4457.
the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 2012;
49 Fernandez LA, Squatrito M, Northcott P, Awan A, Holland EC, Taylor MD,
150:780–791.
et al. Oncogenic YAP promotes radioresistance and genomic instability in
medulloblastoma through IGF2-mediated Akt activation. Oncogene 2012; 74 Gargini R, Escoll M, Garcia E, García-Escudero R, Wandosell F, Antón IM,
31:1923–1937. et al. WIP drives tumor progression through YAP/TAZ-dependent
50 Liu Y, Xin Y, Ye F, Wang W, Lu Q, Kaplan HJ, Dean DC. Taz-tead1 links cell- autonomous cell growth. Cell Rep 2016; 17:1962–1977.
cell contact to zeb1 expression, proliferation, and dedifferentiation in retinal 75 Wang W, Li N, Li X, Tran MK, Han X, Chen J. Tankyrase inhibitors target YAP
pigment epithelial cells. Invest Ophthalmol Vis Sci 2010; 51:3372–3378. by stabilizing angiomotin family proteins. Cell Rep 2015; 13:524–532.
51 Gao Y, Zhang W, Han X, Li F, Wang X, Wang R, et al. YAP inhibits squamous 76 Troilo A, Benson EK, Esposito D, Garibsingh RA, Reddy EP, Aaronson SA.
transdifferentiation of Lkb1-deficient lung adenocarcinoma through Angiomotin stabilization by tankyrase inhibitors antagonizes constitutive
ZEB2-dependent DNp63 repression. Nat Commun 2014; 5:4629. TEAD-dependent transcription and proliferation of human tumor cells with
52 Zhou D, Conrad C, Xia F, Park JS, Payer B, Yin Y, et al. Mst1 and Mst2 Hippo pathway core component mutations. Oncotarget 2016;
maintain hepatocyte quiescence and suppress hepatocellular carcinoma 7:28765–28782.
development through inactivation of the Yap1 oncogene. Cancer Cell 2009; 77 Wang H, Lu B, Castillo J, Zhang Y, McAllister G, Lindeman A, et al. Tankyrase
16:425–438. inhibitor sensitizes lung cancer cells to endothelial growth factor receptor
53 Lu L, Li Y, Kim SM, Bossuyt W, Liu P, Qiu Q, et al. Hippo signaling is a potent (EGFR) inhibition via stabilizing angiomotins and inhibiting YAP signaling.
in vivo growth and tumor suppressor pathway in the mammalian liver. Proc J Biol Chem 2016; 291:15256–15266.
Natl Acad Sci USA 2010; 107:1437–1442. 78 Liu-Chittenden Y, Huang B, Shim JS, Chen Q, Lee SJ, Anders RA, et al.
54 Song H, Mak KK, Topol L, Yun K, Hu J, Garrett L, et al. Mammalian Mst1 and Genetic and pharmacological disruption of the TEAD-YAP complex
Mst2 kinases play essential roles in organ size control and tumor suppresses the oncogenic activity of YAP. Genes Dev 2012;
suppression. Proc Natl Acad Sci USA 2010; 107:1431–1436. 26:1300–1305.
55 Lee DH, Park JO, Kim TS, Kim SK, Kim TH, Kim MC, et al. LATS-YAP/TAZ 79 Panjehpour M, DeNovo RC, Petersen MG, Overholt BF, Bower R,
controls lineage specification by regulating TGFbeta signaling and Rubinchik V, Kelly B. Photodynamic therapy using Verteporfin
Hnf4alpha expression during liver development. Nat Commun 2016; (benzoporphyrin derivative monoacid ring A, BPD-MA) and 630 nm laser
7:11961. light in canine esophagus. Lasers Surg Med 2002; 30:26–30.
56 Cai J, Zhang N, Zheng Y, de Wilde RF, Maitra A, Pan D. The Hippo signaling 80 Lin Z, Guo H, Cao Y, Zohrabian S, Bower R, Rubinchik V, et al. Acetylation of
pathway restricts the oncogenic potential of an intestinal regeneration VGLL4 regulates Hippo-YAP signaling and postnatal cardiac growth. Dev
program. Genes Dev 2010; 24:2383–2388. Cell 2016; 39:466–479.
57 Lee KP, Lee JH, Kim TS, Kim TH, Park H-D, Byun J-S, et al. The Hippo- 81 Zhang W, Gao Y, Li P, Shi Z, Guo T, Li F, et al. VGLL4 functions as a new
Salvador pathway restrains hepatic oval cell proliferation, liver size, and liver tumor suppressor in lung cancer by negatively regulating the YAP-TEAD
tumorigenesis. Proc Natl Acad Sci U S A 2010; 107:8248–8253.
transcriptional complex. Cell Res 2014; 24:331–343.
58 Nishio M, Hamada K, Kawahara K, Sasaki M, Noguchi F, Chiba S, et al. 82 Jiao S, Wang H, Shi Z, Dong A, Zhang W, Song X, et al. A peptide mimicking
Cancer susceptibility and embryonic lethality in Mob1a/1b double-
VGLL4 function acts as a YAP antagonist therapy against gastric cancer.
mutant mice. J Clin Invest 2012; 122:4505–4518.
Cancer Cell 2014; 25:166–180.
59 Wang H, Du YC, Zhou XJ, Liu H, Tang SC. The dual functions of YAP-1 to
83 Pobbati AV, Han X, Hung AW, Weiguang S, Huda N, Chen GY, et al.
promote and inhibit cell growth in human malignancy. Cancer Metastasis Rev
Targeting the central pocket in human transcription factor TEAD as a
2014; 33:173–181.
potential cancer therapeutic strategy. Structure 2015; 23:2076–2086.
60 Barry ER, Morikawa T, Butler BL, Shrestha K, de la Rosa R, Yan KS, et al.
84 Anand R, Maksimoska J, Pagano N, Wong EY, Gimotty PA, Diamond SL,
Restriction of intestinal stem cell expansion and the regenerative
et al. Toward the development of a potent and selective organoruthenium
response by YAP. Nature 2013; 493:106–110.
61 Cottini F, Hideshima T, Xu C, Sattler M, Dori M, Agnelli L, et al. Rescue of mammalian sterile 20 kinase inhibitor. J Med Chem 2009; 52:1602–1611.
Hippo coactivator YAP1 triggers DNA damage-induced apoptosis in 85 Fan F, He Z, Kong LL, Chen Q, Zhang S, Ye J, et al. Pharmacological
hematological cancers. Nat Med 2014; 20:599–606. targeting of kinases MST1 and MST2 augments tissue repair and
62 Schlegelmilch K, Mohseni M, Kirak O, Pruszak J, Rodriguez JR, Zhou D, et al. regeneration. Sci Transl Med 2016; 8:352ra108.
Yap1 acts downstream of alpha-catenin to control epidermal proliferation. 86 Miller E, Yang J, DeRan M, Wu C, Su A, Bonamy GM, et al. Identification of
Cell 2011; 144:782–795. serum-derived sphingosine-1-phosphate as a small molecule
63 Reginensi A, Scott RP, Gregorieff A, Bagherie-Lachidan M, Chung C, regulator of YAP. Chem Biol 2012; 19:955–962.
Lim DS, et al. Yap- and Cdc42-dependent nephrogenesis and 87 Bae JS, Kim SM, Lee H. The Hippo signaling pathway provides novel anti-
morphogenesis during mouse kidney development. PLoS Genet 2013; 9: cancer drug targets. Oncotarget 2017; 8:16084–16098.
e1003380. 88 Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy:
64 Moroishi T, Hayashi T, Pan WW, Fujita Y, Holt MV, Qin J, et al. The Hippo toward combination strategies with curative potential. Cell 2015;
pathway kinases LATS1/2 suppress cancer immunity. Cell 2016; 167:1525. 161:205–214.
e17–1539.e17. 89 Pico de Coana Y, Choudhury A, Kiessling R. Checkpoint blockade for cancer
65 Guo L, Teng L. YAP/TAZ for cancer therapy: opportunities and challenges therapy: revitalizing a suppressed immune system. Trends Mol Med 2015;
(review). Int J Oncol 2015; 46:1444–1452. 21:482–491.

Copyright r 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

You might also like