Hansen Et Al 2015 Hippo Signaling

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Feature Review

YAP and TAZ: a nexus for Hippo


signaling and beyond
Carsten Gram Hansen1,2*, Toshiro Moroishi1,2*, and Kun-Liang Guan1,2
1
Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
2
Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA

The Hippo pathway is a potent regulator of cellular physiological functions mediated by either nuclear or cyto-
proliferation, differentiation, and tissue homeostasis. plasmic localization of YAP/TAZ have been revealed and
Here we review the regulatory mechanisms of the are providing insights into how YAP/TAZ integrates and
Hippo pathway and discuss the function of Yes-associ- modulates the cellular microenvironment.
ated protein (YAP)/transcriptional coactivator with a Drawing on studies in both Drosophila and mammals,
PDZ-binding domain (TAZ), the prime mediators of the we highlight recent findings and provide a current over-
Hippo pathway, in stem cell biology and tissue regen- view of the molecular regulation of the Hippo network in
eration. We highlight their activities in both the nucleus mammals, with a particular focus on the key downstream
and the cytoplasm and discuss their role as a signaling effectors YAP and TAZ. We highlight the role of YAP/TAZ
nexus and integrator of several other prominent sig- as a nexus and integrator for multiple prominent pathways
naling pathways such as the Wnt, G protein-coupled and signaling organelles that play key roles in the control
receptor (GPCR), epidermal growth factor (EGF), bone of cell fate and tissue regeneration.
morphogenetic protein (BMP)/transforming growth
factor beta (TGFb), and Notch pathways. At the core of the Hippo pathway: phosphorylation-
dependent regulation
The Hippo pathway: signal integrator of cell The Hippo pathway comprises a large network of proteins
proliferation and differentiation with the central components of a regulatory kinase module
Coordinated regulation of stem cell proliferation and dif- and a transcriptional module (Figure 1). Many components
ferentiation is essential to maintain tissue mass and ho- of the Hippo pathway are highly conserved from Drosophila
meostasis. The Hippo pathway was initially identified to mammals, including mammalian STE20-like protein
through genetic mosaic screens for suppressors of tissue kinase 1/2 (MST1/2) (Hippo homolog), Salvador family
overgrowth in Drosophila melanogaster [1,2]. Later studies WW domain-containing protein 1 (SAV1) (Salvador homo-
identified that the general components and functions of the log), large tumor suppressor 1/2 (LATS1/2) (Warts homolog),
pathway are highly conserved in mammals [3]. Salient MOB kinase activator 1A/B (MOB1A/B) (Mats homolog),
properties of the Hippo pathway are in regulating cell MAP4K4 (Misshapen homolog), YAP and TAZ (Yorkie
proliferation, survival, and differentiation – all of which homolog), and TEA domain family members 1–4
are particularly important processes in tissue development (TEAD1–4) (Scalloped homolog). Although the core compo-
and regeneration. Deregulation of this pathway is further- nents of the Hippo pathway are extensively conserved, a
more implicated in cancer development [4–6]. divergent role of the upstream regulators through evolution
Downstream effectors of the Hippo kinase cascade are has become apparent (Box 1).
the paralogs and transcriptional coactivators YAP and The Hippo pathway responds to various upstream sti-
TAZ (also known as WWTR1) [3]. The Hippo kinase cas- muli from the cellular microenvironment including me-
cade is the major regulator of YAP/TAZ by phosphorylating chanical signals, cellular stress, extracellular stimuli,
YAP/TAZ and thereby inhibiting their nuclear activities. polarity, and adhesion cues that are integrated through
Recent studies have expanded the complexity of YAP/TAZ upstream regulators. These upstream regulators promote
considerably, identifying new upstream regulatory compo- the activation of the Hippo kinase cascade by activating the
nents and revealing its integration with other signaling mammalian Ser/Thr kinases LATS1/2, which phosphory-
pathways such as the GPCR [7–13], Wnt [14–17], TGFb late and promote either the cytoplasmic retention of YAP
[18–20], EGF [21–24], and Notch [25–28]. In addition to and TAZ via a 14-3-3 interaction or the degradation of YAP/
expanding the number of inputs that modulate YAP/TAZ, TAZ, ultimately preventing TEAD-mediated transcription
the downstream mechanisms of YAP/TAZ have also grown [1,3]. Activation of the upstream kinase module thus con-
in complexity. Besides transcriptional regulation, new cludes in inhibition of the transcriptional output module.
Corresponding author: Guan, K.-L. (kuguan@ucsd.edu).
One of the key questions concerning the Hippo pathway
Keywords: extracellular matrix; stem cells; regeneration; primary cilia; Wnt; Notch. is the molecular mechanism of LATS activation. In a
*
These authors contributed equally to this work. classical view, MST1/2-mediated phosphorylation of
0962-8924/ LATS1/2 at the hydrophobic motif is necessary for LATS
ß 2015 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tcb.2015.05.002 kinase activation [29]. However, the requirement of

Trends in Cell Biology, September 2015, Vol. 25, No. 9 499


Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

Cell-cell contact Cell polarity Extracellular signaling Mechanotransducon Cellular stress

Upstream regulators

Kinase module
MST1/2 MAP4K4 ?
SAV1
P

P P
P LATS1/2 LATS1/2
MOB1A/B

14-3-3
P P P
YAP/TAZ YAP/TAZ YAP/TAZ
Cytoplasmic Degradaon
retenon

CTGF
CYR61
AREG
YAP/TAZ AMOTL2
TEAD TEAD BIRC5...

Transcriponal module

Hippo pathway: ON Hippo pathway: OFF


TRENDS in Cell Biology

Figure 1. The core of the Hippo pathway. The core components of the Hippo pathway comprise a regulatory serine–threonine kinase module and a transcriptional module.
In mammals, this kinase module comprises two groups of kinases: mammalian STE20-like protein kinase 1 (MST1) (also known as STK4) and MST2 (also known as STK3)
and large tumor suppressor 1 (LATS1) and LATS2 in combination with their activating adaptor proteins Salvador family WW domain-containing protein 1 (SAV1) and MOB
kinase activator 1A/B (MOB1A/B), respectively. MAP4K4 has recently been identified as an alternative activator of LATS independent of MST. The transcriptional module
comprises the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with a PDZ-binding domain (TAZ) and several transcription factors,
of which the most prominently studied are TEA domain family members 1–4 (TEAD1–4). In a classical view, when the upstream kinase module is activated, MST1/2
phosphorylates SAV1, MOB1A/B, and LATS1/2 to activate the LATS1/2 kinases, which in turn directly phosphorylate YAP and TAZ on multiple serine residues leading to
cytoplasmic retention of YAP/TAZ via a 14-3-3 interaction. Furthermore, the phosphorylation of YAP/TAZ by LATS1/2 primes YAP/TAZ for proteasomal degradation. The
requirement for MST1/2 to phosphorylate LATS1/2 is cell-type dependent, suggesting the existence of additional kinases (drawn as a question mark) that regulate the
activating phosphorylation of LATS1/2. By contrast, when the kinase module is inactivated, hypophosphorylated YAP/TAZ translocate into the nucleus and induce target
gene expression.

MST1/2 for LATS1/2 phosphorylation is context- and cell enteroblasts had little effect on midgut proliferation. More-
type-dependent. Loss of MST1/2 in mouse embryonic fibro- over, an increase in midgut proliferation resulting from
blasts does not significantly affect LATS1/2 phosphoryla- Misshapen knockdown in enteroblasts was not rescued by
tion and activation [30]. Furthermore, the forced Hippo overexpression, suggesting that Hippo and Mis-
expression of plasma membrane-targeting Warts (the Dro- shapen are not redundant in this cell type [32]. Similarly,
sophila homolog of LATS1/2) partially rescues the eye disc the mammalian Misshapen homolog MAP4K4 activates
phenotype of Hippo (the Drosophila homolog of MST1/2) LATS2, suggesting Misshapen/MAP4K4 is an evolution-
mutant flies, suggesting the existence of additional kinases arily conserved MST1/2-independent activator of LATS1/2,
that regulate the activity-dependent phosphorylation of although it is not entirely clear whether Misshapen/
LATS1/2 or Warts [31]. Indeed, an alternative pathway MAP4K4 directly phosphorylates and activates LATS
activating Warts was identified [32]. In the Drosophila [32]. Taken together, these observations suggest a cell
adult midgut, the protein kinase Misshapen activates type-dependent requirement of Hippo (MST1/2) and Mis-
Warts to negatively regulate the activity of Yorkie shapen (MAP4K4) for Warts (LATS1/2) activation and Yki
and its target gene Upd3 (a JAK–STAT pathway ligand) (YAP/TAZ) inhibition. We speculate that these direct med-
in differentiating enteroblasts, thereby restricting intesti- iators of LATS activation (and thereby YAP and TAZ
nal stem cell division. Notably, depletion of Hippo in inhibition) could be regulated and fine-tuned on cellular
500
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

stimulation, depending on the cellular ratios and differ- [49]. Interestingly, adherens junctions-localized LATS
ential regulation of these distinct LATS-activating kinases phosphorylate AMOT to further stabilize the
kinases. Elucidating this context dependency and the AMOT–LATS interaction [49], establishing a potential
spatiotemporal hierarchy in the dynamics of LATS acti- positive feedback of Hippo pathway activation. By contrast,
vation will reveal further insights into the Hippo pathway. however, when LATS kinases are recruited to adherens
Besides phosphorylation, recent studies have also junctions by the LIM domain-containing protein AJUBA
revealed additional levels of regulation via protein stabili- in response to cytoskeletal tension, LATS-mediated phos-
ty of the Hippo kinase/transcription modules [33–37] and phorylation of YAP and TAZ is impaired [51,52]. Therefore,
by scaffolding the components of the Hippo pathway it is not only LATS localization but also other unknown
[38]. Some of these regulatory mechanisms are highlighted factors that affect the activation state of LATS. Moreover,
in Box 2. This complexity adds an extra layer of divergent another report showed that NF2 inhibits the E3 ubiquitin
regulation of the Hippo pathway. ligase CRL4DCAF1-mediated ubiquitylation of LATS1/2 in
the nucleus [50]. In NF2-mutant tumors, CRL4DCAF1-medi-
Upstream components of the Hippo pathway ated ubiquitylation of LATS1 results in its proteasomal
Spatial regulation of LATS activation degradation, while that of LATS2 leads to loss of its kinase
Epithelial cells adhere mutually through cell–cell junc- activity potentially as a result of conformational change
tions such as tight, adherens, and basolateral, dividing [50]. Together, these observations suggest that NF2 acti-
the plasma membrane into an apical domain and a baso- vates LATS kinases at multiple levels, yet the precise
lateral domain and thereby establishing an apical–basal mechanism remains unclear.
polarity. Studies conducted in both D. melanogaster and
mammals have revealed that many components of cell–cell Physical regulation of YAP and TAZ
junction- and cell polarity-related protein complexes regu- In addition to regulating YAP and TAZ through phosphor-
late the Hippo pathway. Moreover, these regulators of YAP ylation by LATS, other upstream branches of the Hippo
and TAZ are especially important in tissue organogenesis pathway directly inhibit the activity of the transcription
and in preventing cancer progression [39–41]. module by complexing YAP/TAZ, thus sequestering and
While YAP and TAZ are found in the cytosol and the tight preventing their nuclear access (Figure 2B). These
junctions in dense cells, these transcriptional coactivators branches include AMOT [53–56], protein tyrosine phos-
translocate to the nucleus and activate TEAD- phatase 14 (PTPN14) [57,58], and a-catenin [59]. The
mediated transcription in sparse cells [42,43]. This regula- AMOT–YAP/TAZ interaction is mediated via PPxY motifs
tion is mediated primarily via homophilic cell–cell contacts of AMOT and WW domains of YAP and TAZ and thus does
that activate the kinase module [42,44]. In addition, the not require YAP and TAZ phosphorylation by the kinase
basolateral junction proteins, including mammalian Scrib- module [53–55]. This regulatory interaction between PPxY
ble (SCRIB) facilitate the activation of the core kinases MST motifs and WW domains is widespread throughout the
and LATS, thereby inhibiting the nuclear activities of YAP/ Hippo pathway [60]. PTPN14 also utilizes its PPxY motifs
TAZ in mammary epithelial cells [45] (Figure 2A). Loss of to directly interact with WW domains of YAP, causing
cell polarity via induction of the epithelial–mesenchymal cytoplasmic localization of YAP and decreased YAP activi-
transition (EMT) causes SCRIB delocalization and TAZ ty [57,58]. The adherens junction protein a-catenin also
activation, which confers self-renewal and tumor-initiation regulates YAP by sequestering YAP–14-3-3 protein com-
capacities on breast cancer stem cells [45]. Furthermore, the plexes in the cytoplasm [59]. However, unlike AMOT, the
apical membrane-associated FERM-domain protein neuro- kinase module that mediates YAP phosphorylation is re-
fibromatosis 2 (NF2) and the WW and C2 domain-contain- quired for the interaction between a-catenin and YAP, as
ing protein kidney and brain expressed protein (KIBRA) 14-3-3 binds to phosphorylated YAP only [3]. Notably, a-
(also known as WWC1) form a complex to activate the core catenin also provides a scaffold for the LATS activation/
kinase cassette [46–48]. NF2 is a potent tumor suppressor, inactivation process (Figure 2A).
mutations of which are associated with neurofibromatosis AMOT is a filamentous actin (F-actin)-binding protein
type II [5], and is a potent activator of the core Hippo kinase that predominantly localizes at tight junctions and F-actin
module [1]. Recent studies have revealed the complexity and competes with YAP for binding to AMOT [61]. Interestingly,
multilevel regulation of LATS kinases by NF2 (Figure 2A). recent studies revealed that LATS1/2 phosphorylate AMOT
NF2 interacts with numerous proteins that are localized at within a conserved F-actin-binding domain [49,61–63],
the plasma membrane [31], including a-catenin and Angio- which inhibits AMOT binding to F-actin thereby potentiat-
motin (AMOT) at adherens junctions [49], but also localizes ing its inhibitory effects on YAP and TAZ activity
in the nucleus [50]. At the plasma membrane of cultured [49,61,62]. LATS kinases can therefore inhibit nuclear
cells, NF2 was shown to recruit LATS kinases and coordi- YAP and TAZ activity both directly and indirectly. Most
nate their activating phosphorylation by the MST–SAV1 studies support an inhibitory role of AMOT on YAP/TAZ
kinase complex [31], highlighting the plasma membrane as [35,49,53–55,61,62], but a recent report indicated that
a critical subcellular compartment for Hippo signal trans- AMOT binds to YAP to support YAP–TEAD mediated
duction. At adherens junctions in the inner cells of preim- transcription in the nucleus and thereby stimulate YAP-
plantation embryos, NF2 directly interacts with a-catenin mediated hepatic tumorigenesis in NF2-deficient mice [64].
and AMOT to recruit LATS kinases, which in turn activates Additional regulatory mechanisms of YAP/TAZ activity,
the kinase module of the Hippo pathway and thereby reg- divergent from its main regulation via sequestration, have
ulates lineage specification in preimplantation embryos also become apparent (Figure 2B). WW domain-binding
501
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

(A) (B)

NF2 KIBRA SAV1


LATS1/2 MST1/2
P
Acvaon AMOT
YAP/TAZ AMOT
P
Tight juncon F-acn
LATS1/2
P
AMOT 14-3-3
Acvaon P
NF2 YAP/TAZ
α -catenin α-catenin
AJUBA
LATS1/2
Adherens juncon Adherens juncon PTPN14
YAP/TAZ
Inacvaon

P
LATS1/2
SCRIB Acvaon
MST1/2
NF2
Basolateral juncon WBP2
MASK
CRL4DCAF1
VGLL4
Ub Ub
LATS1/2 Ub
Ub Ub VGLL4 YAP/TAZ
Ub
Inacvaon Ub TEAD TEAD
LATS1/2

Nucleus Nucleus

TRENDS in Cell Biology

Figure 2. Regulators of Yes-associated protein (YAP)/transcriptional coactivator with a PDZ-binding domain (TAZ) activities. (A) Spatial organization and regulation of the
kinase module. Neurofibromatosis 2 (NF2) (also known as Merlin) activates large tumor suppressor (LATS) kinases at multiple levels. NF2 recruits LATS at the plasma
membrane to coordinate the activating phosphorylation by the mammalian STE20-like protein kinase (MST)–Salvador family WW domain-containing protein 1 (SAV1)
kinase complex. NF2 directly interacts with a-catenin and Angiomotin (AMOT) to recruit LATS kinases at the adherens junction, which in turn activate LATS. NF2 inhibits the
E3 ubiquitin ligase CRL4DCAF1-mediated ubiquitylation of LATS1/2 in the nucleus to inhibit proteasomal degradation of LATS1 and ubiquitylation-induced conformational
change of LATS2. Scribble (SCRIB) recruits the core kinases MST and LATS to facilitate LATS activation at the basolateral junction. LATS recruitment by AJUBA at adherens
junctions inhibits LATS kinase activity. (B) Physical regulation of the transcription module. AMOT, protein tyrosine phosphatase 14 (PTPN14), and a-catenin inhibit the
activity of the transcription module by complexing YAP and TAZ, thus sequestering and preventing their nuclear access. AMOT interacts with and thereby causes
cytoplasmic sequestration of YAP and TAZ by recruiting them to tight junctions. PTPN14 also causes cytoplasmic localization of YAP and decreased nuclear YAP activity.
The adherens junctions protein a-catenin also regulates YAP by sequestering YAP–14-3-3 protein complexes in the cytoplasm. WW domain-binding protein 2 (WBP2) and
multiple ankyrin repeats single KH domain-containing protein (MASK) interact with nuclear YAP and TAZ, which enhances YAP and TAZ transcriptional coactivator
properties, but the precise mechanism remains unknown. Vestigial-like family member 4 (VGLL4) represses target gene expression by competing with YAP–TEA domain
family (TEAD) interactions.

protein 2 (WBP2) interacts with YAP and TAZ in a WW is downregulated in lung [70] and gastric [71] cancers and
domain- and PPxY motif-dependent manner, enhancing high MASK1 expression is associated with poor prognosis
YAP and TAZ transcriptional coactivator properties for breast cancer patients [67].
[65,66]. Suppression of WBP2 expression prevents the
overgrowth phenotype of Warts mutant flies and inhibits Nuclear functions of YAP and TAZ
TAZ-driven transformation of MCF10A mammary epithe- The most widely studied role of YAP and TAZ is as
lial cells [65,66]. Multiple ankyrin repeats single KH do- transcriptional coactivators. YAP and TAZ do not contain
main-containing protein (MASK) has also been shown any DNA-binding domains and thus bind to and elicit
to potentiate YAP activity through direct binding their transcription activation via cognate TFs (Figure 3A).
[67,68]. Recent data suggest that vestigial-like family Accumulating evidence shows that the TEAD family of TFs
member 4 (VGLL4) is a natural inhibitor of YAP–TEAD are major mediators of YAP and TAZ nuclear activity
interaction that represses target gene expression and therefore gene output [40,43,70,72,73] (Figure 1).
[69,70]. The Tondu domains of VGLL4 interact with TEAD Nonetheless, YAP and TAZ do utilize other TFs, and some
transcription factors (TFs) and may compete for common contextual transcriptional responses are therefore mediated
binding sites with YAP and TAZ [69,70]. The physiological/ by additional TFs such as SMADs, p63, RUNX, and PAX
pathological roles of these regulations are primarily [3,74], which diversifies the transcriptional output of the
characterized in YAP/TAZ-induced tumorigenesis. VGLL4 Hippo pathway.
502
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

(A) (B)
Transcriponal co-acvaon miRNA processing
P
YAP/TAZ

Transcripon factors
YAP/TAZ
RUNX SWI/SNF chroman Histone methyltransferase DDX17 DDX17
remodeling complex complex
p73
Microprocessor
p63 BRM complex
NCOA6
SMAD
PAX
YAP/TAZ Target gene
pri-miRNA pre-miRNA
TEAD transcripon

(C) Wnt signal P


(D) TGFβ/BMP–SMAD signal
YAP/TAZ

Proteasomal P β-catenin
P
degradaon YAP/TAZ
YAP/TAZ
DVL
SMAD2/3
?
SHP2
P
P YAP/TAZ
YAP/TAZ
P
Destrucon complex CDC73

PAF complex

P SHP2
YAP/TAZ
CDC73
β-catenin YAP/TAZ YAP
Target gene Target gene
TCF transcripon SMAD2/3 SMAD1 transcripon

TRENDS in Cell Biology

Figure 3. Function of nuclear and cytoplasmic Yes-associated protein (YAP) and transcriptional coactivator with a PDZ-binding domain (TAZ). (A) Transcriptional
coactivation. Nuclear YAP and TAZ bind to transcription factors (TFs) and recruit the NCOA6 methyltransferase complex as well as the BRM-containing SWI/SNF chromatin-
remodeling complex, which mediates TF target-specific gene transcription. (B) miRNA processing. Nuclear YAP inhibits DDX17, a regulatory component of the
microprocessor complex. This nuclear inhibition decreases global miRNA on YAP stimulation. Of note, some specific miRNAs are upregulated on YAP nuclear activation.
(C) Wnt signaling. Cytoplasmic YAP and TAZ sequester b-catenin and in some instances recruit b-TrCP to the destruction complex. This leads to nuclear b-catenin
downregulation and therefore dampens Wnt signaling. Cytoplasmic YAP and TAZ also negatively regulate Wnt signaling by sequestering b-catenin and disheveled (DVL).
YAP and TAZ dictate nuclear and cytoplasmic localization of SHP2 and therefore its function. This feeds into the Wnt signaling pathway because nuclear SHP2
dephosphorylates parafibromin (CDC73), which leads to increased b-catenin transcriptional activity. (D) Transforming growth factor beta (TGFb)/BMP–SMAD signal.
Cytoplasmic YAP and TAZ sequester SMAD2/3 in the cytoplasm and therefore inhibit SMAD2/3-mediated signaling, although these mechanisms seem not to be universal
and somewhat controversial. Nuclear YAP activates SMAD1-mediated transcription.

A functional explanation of how YAP and TAZ mechanis- biogenesis of certain miRNAs, such as miR-16, -21, -23a,
tically regulate TEAD transcriptional activity has been -29, -107, and -152, by increasing Dicer via the Let-7 family
revealed [75–78]. TAZ (and YAP) interacts with components of miRNAs [82,83]. YAP and TAZ can thus function as both
of the SWI/SNF chromatin-remodeling complex [77,78] and positive and negative mediators of miRNA processing,
YAP (and TAZ) recruits a methyl transferase complex into depending on their targets.
proximity with the TF, which leads to increased histone
(H3K4) methylation and transcription of target genes YAP/TAZ as a signaling network
[75,76]. These complex formations are functionally mediat- The Hippo pathway does not appear to have a unique
ed via interactions between WW domains of YAP/TAZ and extracellular ‘Hippo pathway-specific’ receptor/ligand in-
PPxY motifs of components of chromatin-remodeling/ teraction that exclusively regulates this pathway. Instead,
methyltransferase complexes [75–78] (Figure 3A). It will it is integrated with a network of other prominent signal-
be interesting to learn whether the same mechanism holds ing pathways that impinge and rely on the Hippo pathway
true for other YAP/TAZ-utilized TFs. In some instances for their comprehensive functions. Moreover, YAP/TAZ
YAP/TAZ also operate as transcriptional corepressors for serve as a cellular manifold and directly mediate transcrip-
additional target genes [79,80]. Intriguingly this nuclear tion of several of these receptors and ligands that when
function also requires TEAD TFs [80]. expressed activate YAP/TAZ signaling. As the activity of
In addition, nuclear YAP has also been found to regulate constitutive positive feedback loops ultimately would lead
miRNA processing. Nuclear YAP/TAZ bind and sequester to futile cycles and to pathological conditions, these must
DEAD box helicase 17 (DDX17) (also known as p72) to be tightly regulated, but how unrestrained YAP and TAZ
repress its association with Microprocessor, a complex that activity is sensed and contained at the cellular level is
regulates miRNA processing (Figure 3B). Therefore, the currently not well understood.
activation of YAP/TAZ in the nucleus is also a general Besides their widely recognized role in cancer [4], the
mechanism of impaired global miRNA biogenesis [81]. physiological importance of YAP/TAZ has also been
Interestingly, nuclear YAP/TAZ conversely stimulate the established in regeneration, development, and stem cell
503
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

biology. We focus on a few tissues and biological contexts intestinal YAP/TAZ nuclear hyperactivity [27,93,95,96].
where the Hippo pathway has been most extensively studied The utilization of an in vivo intestine-specific gene trans-
and discuss these findings within the framework of other fer technique revealed a dual role for YAP/TAZ in renewal
signaling pathways and cellular signaling organelles. of the intestinal epithelium. In this process YAP/TAZ use
two distinct TFs [96]: TEAD [43,72,73] for stem and
YAP/TAZ and Wnt signaling in the intestine progenitor cell proliferation and Krüppel-like factor 4
YAP/TAZ, the Hippo Pathway, and Wnt signaling are (Klf4) for differentiation into goblet cells [96].
profoundly integrated [84]. In the absence of a Wnt signal, While these findings suggest a proliferative role of YAP
b-catenin remains phosphorylated, resulting in its degra- in intestinal homeostasis, other reports observed an
dation and the transcriptional inhibition of Wnt target inhibitory effect of YAP in intestinal regeneration and
genes [85]. The cytoplasmic retention of YAP/TAZ via its Wnt-driven hyperplasia [16,86]. Moreover, known target
phosphorylation results in the sequestration of b-catenin genes of YAP/TAZ–TEAD include BMP4 and DKK1
in the cytoplasm [86] and in some instances the recruit- [97,98], which are negative regulators of Wnt signaling
ment of b-TrCP to this complex, leading to the degradation and crypt formation [85,97,99] (Figure 4). Future studies
of b-catenin [14,87] (Figure 3C). In addition, cytoplasmic should examine how other pathways that are known to be
YAP/TAZ interact with disheveled (DVL) to inhibit differentially induced on challenge in the intestine [100],
Wnt target gene transcription [15,16]. The tyrosine phos- such as inflammation [101], integrate these responses via
phatase SHP2 (encoded by the PTPN11 gene), like YAP Lgr5, Wnt, and the Hippo pathway.
and TAZ [42], shuttles between the nucleus and the cyto-
plasm where its nuclear localization promotes b-catenin- TGFb and BMP signaling
dependent transcription [88,89]. SHP2 is excluded from The TGFb superfamily is a group of secreted morphogens
the nucleus in high-density cells by its interaction with that are potent regulators of stem cell pluripotency and
phosphorylated YAP and TAZ [89], suggesting that cyto- development [102,103]. TGFb binds to its cognate serine–
plasmic YAP/TAZ functionally terminate Wnt signaling threonine kinase receptor to induce phosphorylation and
through at least three separate mechanisms [15,16,87]. activation of SMAD TFs [102]. The Hippo pathway is
By contrast, nuclear YAP/TAZ promotes nuclear b-catenin, integrated with TGFb and BMP signaling on multiple
which mediates T cell factor (TCF)-dependent transcription levels [18–20,79,97,104] (Figures 3D and 4).
(Figure 3C). Nuclear YAP/TAZ also facilitate nuclear In human embryonic stem cells, impairment of the nu-
translocation of SHP2, which in turn dephosphorylates clear translocation of TAZ results in the sequestration of a
CDC73 (also known as parafibromin) to stimulate complex comprising TAZ, SMAD2, and SMAD3 in the cyto-
CDC73–b-catenin binding and ultimately activate Wnt tar- sol [19] (Figure 3D). However, this cytoplasmic function is
get gene transcription [88–90], providing additional func- not universal, as conflicting data have recently been pub-
tional links between Wnt and Hippo signaling (Figure 3C). A lished in a range of cell lines [105,106]. On activation,
negative regulator of both b-catenin and YAP was also nuclear YAP/TAZ form a complex with SMAD2/3 to bind
recently identified. Protein kinase C zeta (PKCz) was iden- TEAD TFs [18] or OCT4, which mediates pluripotency
tified to phosphorylate and thereby inhibit both b-catenin [79]. On loss of YAP/TAZ, these stem cells lose pluripotency
and YAP, highlighting the atypical PKCs as mediators of potential and differentiate [19,79,107]. In addition, nuclear
intestinal homeostasis and regeneration by modulating YAP supports SMAD1 target gene expression downstream
both YAP and b-catenin activity [91]. A general picture of BMP [108]. YAP plays a role in regulating both embryonic
therefore emerges whereby nuclear YAP/TAZ potentiates and adult lung progenitor cell differentiation by directly
Wnt signaling whereas cytoplasmic YAP/TAZ, due to an specifying the epithelial cell lineage [109,110]. YAP defi-
activated Hippo pathway, inhibits Wnt signaling [15,16,92]. ciency leads to epithelial progenitors that are unable to
In addition, YAP has also been reported to be a b-catenin properly respond to local TGFb-induced cues [110], result-
target gene [17] and thus, in this instance, active Wnt ing in the differentiation of progenitors into epithelial air-
signaling increases YAP abundance and hence transcrip- way cells [109]. Moreover, increased nuclear YAP activity
tional activity, suggesting that Wnt target genes can also leads to dedifferentiation of secretory cells into basal-like
feed back on the Hippo pathway and potentiate its signaling. cells, which is directly mediated via a YAP–p63 interaction
The close reciprocal and complex relationship between [109]. However, it remains to be determined whether YAP/
the Hippo pathway and Wnt has been well studied in the TAZ-mediated, TGFb-dependent transcription is regulated
intestine [16,93]. Indeed, YAP/TAZ expression is predom- exclusively via SMADs [104] or whether it may also require
inantly localized to the bottom of crypts where Lgr5+ TEADs [18,79] or context-dependent partnering with
intestinal stem cells can be found [16,27,85,87]. While diverse TFs as has been recently observed. It is a task for
YAP/TAZ are not essential for intestinal homeostasis the future to discover exactly what is regulated via SMADs,
[87,94], they are required under stress conditions where TEADs, and potentially other TFs and how these diverse
intestinal proliferation is necessary for survival, such as in complexes are regulated.
the regeneration of damaged tissue after dextran sodium YAP/TAZ and ligands of the BMP/TGFb family can also
sulfate treatment [93]. In addition, nuclear YAP/TAZ are create a positive feedback loop that controls the Hippo
important mediators of adenomatous polyposis coli (APC)- pathway [20,97,111–113]. TAZ induces BMP4 transcrip-
mediated hyperplasia due to a combination of both in- tion and promotes cell migration revealing a TAZ/TEAD/
creased nuclear b-catenin and YAP/TAZ signaling [87,93], BMP signaling axis in mammary epithelial cells [97]. In
a phenotype that can be mimicked by genetically inducing addition, integrin-linked kinase, an integrin-associated
504
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

NRG1 BMP
Wnt GPCR EGF TGFβ

Gαq/11 Gα12/13 Gαs

Signal crosstalk

DKK1 NOTCH2

BMP4 ITGB2
LATS1/2
AREG LAMA5

JAG1 ZYX

β-catenin YAP YAP/TAZ ICD-ERBB4 YAP YAP/TAZ


TBX5 TEAD TEAD TEAD SMAD2/3

TRENDS in Cell Biology

Figure 4. Signal crosstalk centering on Yes-associated protein (YAP) and transcriptional coactivator with a PDZ-binding domain (TAZ). Wnt signaling activates YAP and TAZ
by inhibiting the formation of the b-catenin destruction complex that in some instances functions as a cytoplasmic sink and causes degradation of YAP and TAZ. Nuclear
YAP can form a complex with b-catenin and the transcription factor TBX5 to promote antiapoptotic gene expression, including BCL2L1 and BIRC5. G protein-coupled
receptor (GPCR) signaling is relayed via small G proteins. Activation of Ga12/13 and Gaq/11 induces YAP and TAZ nuclear translocation and activity, whereas active Gas
represses YAP and TAZ nuclear activity, which causes either activation or inhibition of large tumor suppressor (LATS) kinase activity. The family of epidermal growth factor
(EGF) ligands activates YAP and TAZ in both a LATS-dependent and -independent manner. EGF receptor (EGFR) stimulation inhibits the kinase module by dissociating
phosphoinositide-dependent kinase-1 (PDK1) from the Hippo pathway components [LATS, mammalian STE20-like protein kinase 1 (MST), and Salvador family WW
domain-containing protein 1 (SAV1)], preventing activation of LATS by MST. EGFR–Ras–MAPK signaling also enhances the AJUBA family protein Wilms’ tumor protein 1-
interacting protein (WTIP) binding to LATS, which inhibits LATS activity toward YAP. In addition, a cytoplasmic intracellular domain of the ERRB4 receptor (ICD-ERBB4) is
cleaved and released on neuregulin 1 (NRG1) stimulation, which in turn binds to YAP and promotes target gene expression. On transforming growth factor beta (TGFb)
signal activation, nuclear YAP and TAZ form a complex with SMAD2, SMAD3, and TEA domain family (TEAD) transcription factors, coordinating a protumorigenic
transcriptional program. YAP and TAZ also directly promote transcription of several receptors and ligands to mediate endocrine and paracrine signal crosstalk and potential
feedback loops. Notable examples are the Wnt signal inhibitor DKK1, ligands of the BMP/TGFb family BMP4, the EGF-like growth factor AREG, NOTCH2, the Notch ligand
Jagged1 (JAG1), integrin/extracellular matrix (ECM) signaling components such as b2-integrin (ITGB2) and the a5 subunit of laminin 511 (LAMA5), and the focal adhesion
component zyxin (ZYX).

actin and tubulin cytoskeleton-interacting effector that lysophosphatidic acid (LPA) and sphingosine 1-phosphate
mediates TGFb signaling, was found to be a negative (S1P) work through GPCRs [7,9] via Ga12/13 and are potent
regulator of the Hippo kinase cascade in cancer cells by inhibitors of the Hippo kinase module and thereby activa-
inhibiting MYPT1, which leads to inhibition of NF2 and tors of nuclear YAP/TAZ (Figure 4). By contrast, ligands
nuclear accumulation of YAP/TAZ [21]. such as epinephrine work via GaS-coupled GPCRs to inhibit
YAP/TAZ through PKA-mediated activation of LATS
GPCR signaling kinases [114,115]. PKA is also implicated as a widespread
A prominent role for GPCRs and their cognate ligands as activator of LATS on other physiological stimuli
regulators of Hippo signaling has been established [7– [114]. GPCR-driven pathologies can be mediated via dysre-
10]. GPCRs are coupled to small heterotrimeric G proteins gulated YAP/TAZ [7,11–13,116] and examples of FDA-
and YAP/TAZ nuclear activities are either activated or approved drugs targeting GPCRs modifying YAP/TAZ
inhibited depending on the G proteins coupled to the active activity are known [10,117]. Several notable GPCR li-
receptors. Notable ligands such as the bioactive lipids gands play crucial roles in intestinal regeneration [118],
505
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

development [119], and stem cell pluripotency [120,121] – Reciprocal interaction with the microenvironment,
biological processes where YAP and TAZ are known to be cellular tension, and the cytoskeleton
key mediators [6,74,107]. Importantly, S1P–YAP signal- Cell shape, cell–cell interactions, mechanotransduction,
ing was shown to be important for cardiac precursors in and matrix adhesiveness and stiffness are all prudent med-
zebrafish [119], but whether these effects by LPA, S1P, iators of cellular state, identity, and niche [135–139]. The
and other GPCR ligands in general are mediated via forces elicited by these interactions are dynamically regu-
activation of YAP/TAZ transcriptional activity in vivo lated and are especially important during processes where
in nonpathological conditions is currently not well under- the cell experiences drastic changes, such as during devel-
stood. It is worthwhile to expand these correlative opment, cell differentiation, and regeneration [74,135–138]
relationships with loss-of-function experiments in mam- – all processes where the Hippo pathway has been shown to
malian animal models. be an important mediator [3,74,84,137,140]. RHO activity
and the actin cytoskeletal organization are robust regula-
EGF family ligands regulate YAP and TAZ tors of YAP/TAZ activity [52,139,141–145]. YAP/TAZ loca-
YAP and TAZ are activated by the EGF family of ligands lizes to the nucleus and is transcriptionally active in cells
(Figure 4). Studies in mammalian systems have so far grown on high-rigidity substrate, under static or cyclic high
focused on cell culture systems, where YAP/TAZ are facil- tension, or in cells lacking cell–cell contact [139–
itators of EGF-mediated cell migration and transcription 142,146,147]. The regulation of YAP/TAZ by these biological
[22–24]. The regulation of YAP/TAZ via EGF-like mole- cues has been shown to impact lineage commitment [148], of
cules is twofold. EGF inhibits the kinase module via which directing mesenchymal stem cell (MSC) fate is best
3-phosphoinositide dependent protein kinase 1 (PDPK1) described. High tension induced by cell spreading or by
and AJUBA proteins, which leads to impairment of SAV1’s culturing on a hard surface promotes the nuclear localiza-
role in the kinase complex [22,23]. In addition, a cyto- tion of YAP/TAZ and drives MSCs toward the osteoblast
plasmic intracellular domain (ICD) of ERBB4 is cleaved lineage. By contrast, growth on a soft surface retains YAP/
and released on neuregulin 1 (NRG1) stimulation that in TAZ in the cytoplasm and promotes adipogenesis, a pheno-
turn binds to and facilitates nuclear translocation of YAP. type that can be recapitulated by depletion of TAZ
This effect is independent of LATS activity [24,122,123] [142,147,149]. YAP/TAZ also induce the expression of a
(Figure 4). Moreover, activation of YAP/TAZ results in the range of cellular tension regulators such as zyxin (a compo-
transcription of the EGF-like growth factor AREG [21], nent of focal adhesions) and the matricellular protein
providing crosstalk between Hippo and EGF signaling CYR61, as well as integrins and their ligands
[21–23]. The biological role of the EGF-like molecules in [43,113,136,146,150–152]. YAP/TAZ thereby serve as nucle-
the regulation of YAP/TAZ is currently not well understood ar relays [135] of mechanical signals exerted by extracellu-
in vivo. As they are prudent modulators of cancer etiology lar matrix (ECM) rigidity, tension, and cell shape.
[124] and stem cell biology [103], it will be interesting to These same integrin/ECM signaling proteins can also
delineate precisely what is mediated via the Hippo pathway. influence YAP/TAZ regulation [20,43,146,151,152]. For ex-
ample, in skeletal stem cells the membrane-anchored
Notch signaling matrix metallopeptidase 14 (MMP14) was found to acti-
A direct link between Notch signaling and YAP has be- vate the b1-integrin/RHO GTPase signaling cascade,
come apparent in several systems, such as the developing which triggered the nuclear accumulation of YAP/TAZ
embryo [28,41,56,125–128], neuronal stem cells [25], and promoted skeletal stem cell lineage commitment [150].
mature hepatocytes [26], and hepatocyte-derived stem
cells [129]. The ligand Jagged1 (JAG1) [26] and the recep- The Hippo pathway and signaling organelles
tor NOTCH2, both known to be critical determinants of Cellular organelles that serve as signaling hubs are im-
biliary cell fate [130,131], are direct YAP–TEAD target portant integrators of YAP/TAZ activity. The Hippo path-
genes [26,27,126,129] (Figure 4). Inducing YAP activity way both controls and is regulated by the primary cilia
in vivo can reprogram hepatocytes via NOTCH activation [94,153–155], a microtubule-based solitary sensory signal-
into progenitor cells [27,129]. ing organelle comprising a special part of the plasma
Notch signaling is also a well-known regulator of satel- membrane in most eukaryotic quiescent cells [156]. The
lite cells [132]. In skeletal muscle, YAP maintains satellite primary cilia respond to mechanical signals [157] and
cells undifferentiated via TEAD, partly by repressing the dynamically localize and regulate a range of receptors
pro-differentiating gene transcription program mediated mediated by YAP/TAZ, such as GPCRs, sonic hedgehog
by MYOD1 and MEF2 [111,133,134]. However, it remains (shh), the TGFb family, receptor tyrosine kinases, and Wnt
to be elucidated whether YAP mediates this effect in receptors [7,10,14,17,22,23,158]. This regulation of recep-
satellite cells directly via NOTCH. tors is predominantly mediated through the cilia transition
Hippo and NOTCH signaling play an important role zone, the cellular gatekeeper for receptors destined for
during mouse embryogenesis from the morula to the blas- cilial localization [156]. Members of the nephrocystin
tocyst stage when the trophectoderm (TE), which gives rise (NPHP) family, established primary cilia-associated pro-
to the placenta, and the inner cell mass (ICM), which gives teins [159], induce TAZ (and YAP) activity. NPHP4 binds
rise to the embryo proper, lineages are specified. NOTCH– to LATS and negatively regulates LATS activity, whereas
RBPJ and YAP–TEAD4 combinatorially bind to distinct NPHP3 and NPHP9 bind directly to TAZ and potentially
promoter regions that directly regulate the transcription of compete with 14-3-3 binding to relieve cytoplasmic reten-
the TE lineage determining TF CDX2 [28,41,56,125–128]. tion [160,161]. Therefore, members of the NPHP family
506
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

increase transcriptional activity of YAP and TAZ binds to the inhibitory transmembrane protein Patched
[153]. Furthermore, TAZ binds the primary cilia-associat- and thereby relieves the GPCR-like molecule Smoothened
ed protein GLIS family zinc finger 3 (Glis3), facilitating from its inhibition [163], an effect that could be mediated
Glis3-mediated transcription [154]. In addition, SHH, the via GPCR signaling. The upstream Hippo kinase module
prototypical primary cilia signaling ligand [162], activates likewise integrates with the primary cilia, as MST and
YAP transcription, abundance, and activity [158]. SHH SAV1 (Figure 1) inhibit the disassembly of primary cilia by

Box 1. Evolutionary divergence of the Hippo pathway


The general structure of the Hippo pathway was drafted by studies while YAP and TAZ are targeted for degradation in mammals, the
conducted in Drosophila melanogaster (reviewed in [1]) and a wealth phosphodegron responsible for this degradation is not conserved in
of studies in both flies and mammals since then has considerably Yorkie [170].
expanded the realization of the Hippo pathway network as an A plausible explanation for this divergence is an evolutionary shift
evolutionarily conserved growth- and cell fate-controlling integrator. at the base of the arthropod lineage [186]. In this transition, fly Fat
The core architecture of the Hippo pathway is highly conserved in gained novel domains that are required for restricting Dachsous to
evolution and homologous components are found in most major regulate the levels of Warts (the Drosophila homolog of LATS1/2)
animal phyla, predating the origin of the Metazoa [175,176] (Figure I). [186]. Unlike fly Fat, murine Fat4 does not appear to regulate the
Multiple upstream branches of the Hippo pathway also regulate the Hippo pathway [186]. Nevertheless, an additional mechanism linking
core components. In D. melanogaster, Tao [thousand and one amino murine Fat4 to Yap is possible because a genetic interaction between
acid protein kinases (TAOKs) in mammals] phosphorylates Hippo (the Fat4 and Yap in mouse embryonic neuroepithelium has been
Drosophila homolog of MST1/2) to activate its kinase activity reported [187]. In mammals, CRBs (crumbs family members) polarity
[177,178], Par-1 [MAP/microtubule affinity-regulating kinases complex forms a complex with AMOT [104], which binds and
(MARKs) in mammals] phosphorylates Hippo to restrict its activity sequesters YAP and TAZ through its PPxY motif [53–55]. AMOT is
[179], Sik [salt-inducible kinases (SIKs) in mammals] phosphorylates likely to have been present in the last common ancestor of all
Salvador (the Drosophila homolog of SAV1) to inhibit its association bilaterian animals but was lost during arthropod evolution [186]. By
with the core kinase module [180], and RAS association domain- contrast, the fly Crumbs-binding protein Expanded [188–190] gained
containing family protein (RASSF) competes with Salvador for Hippo a novel C-terminal domain that contains a PPxY motif and is thus
and recruits a Drosophila PP2A phosphatase complex (dSTRIPAK) to able to interact with Yorkie [186,191,192]. The Crumbs complex
dephosphorylate and inactivate Hippo [181,182]. However, except for thereby exchanges AMOT for Expanded at the base of the arthropod
TAOK [177,178], the functions of these upstream regulators do not lineage. Expanded also regulates the Hippo pathway by forming a
appear to be conserved in mammals: MARK acts downstream of liver complex with Merlin (NF2 in mammals) and Kibra in flies
kinase B1 (LKB1) to activate the core kinase module through SCRIB [48,193]. Therefore, although groundbreaking discoveries continue
[183]; the phosphorylation site in Salvador by Sik is not conserved in to be pioneered in the fly fundamental differences exist in the
human SAV1 [180]; and multiple RASSF isoforms in mammals have regulatory mechanisms of core Hippo components between flies and
different roles in the mammalian Hippo pathway [184,185]. Also, mammals.

Drosophila Mammals
Crbumbs

CRBs
Fat

Scribble SCRIB
Yorkie Expanded YAP/TAZ AMOT

Merlin NF2
dSTRIPAK
Dachsous Kibra KIBRA
dRASSF Par-1 RASSF MARK
Tao TAOK

Sik SIK

Misshapen Hippo MAP4K4 MST1/2


14-3-3 14-3-3
Salvador P SAV1 P
Yorkie YAP/TAZ
Core components

Warts LATS1/2
Mats MOB1A/B

Degradaon
AMOT
Yorkie YAP/TAZ YAP/TAZ
Target gene transcripon Target gene transcripon
Scalloped TEAD

TRENDS in Cell Biology

Figure I. Evolutionary divergence of the Hippo pathway.

507
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

MST-mediated phosphorylation of AURKA, an important interactome studies also link endosomal compartments to
regulator of the primary cilia disassembly complex, and by the Hippo pathway [165–167]. Yorkie (the fly YAP/TAZ
association with the NPHP transition-zone complex, there- homolog, see Box 1) is recruited onto endolysosomes via
by inducing an increase in steady-state primary cilial Leash, a member of the arrestin family, which leads to
abundance [153]. A range of diseases caused by dysfunc- degradation and inhibition of Yorkie nuclear activity
tional primary cilia are termed ciliopathies [156,164] and, [165]. Overexpression of Warts induces Yorkie-contain-
intriguingly, TAZ knockout animals develop polycystic ing vesicular structures, suggesting that phosphorylation
kidneys [94] and express lower levels of multiple genes of Yorkie facilitates its vesicular localization [165].
important for primary cilial morphogenesis; thus, these Although it remains unclear whether similar regulation
mice have malfunctioning cilia and exhibit a phenotype of YAP and TAZ occurs in mammalian systems, a role for
reminiscent of known human ciliopathies [94,164]. By con- the autophagosome/lysosome system in YAP protein sta-
trast, TAZ depletion in tissue culture cells such as RPE1 bility in mice has been observed [169]. YAP accumulates
cells induce ciliogenesis [155]. It is worth noting that the in the kidney of tuberous sclerosis complex 1 (TSC1)-
primary cilia are turned over at each cell cycle and that deficient mice due to impaired autophagy and thereby
YAP/TAZ causes cell cycle progression; however, since the promotes abnormal cell proliferation and polycystosis,
primary cilia are present in quiescent cells only, it needs to reminiscent of human perivascular epithelioid cell
be fully determined which of these effects are causative or tumors from TSC patients. How lysosomal regulation of
correlative. YAP can be reconciled with previous reports [170] that
The endolysosomal system is also a mediator of the indicate proteasomal degradation of YAP is currently
Hippo pathway [165–167]. Through a genetic screen for unclear. However, these findings suggest an intriguing
conditional growth suppressor mutants, Myopic [the new mode of regulation, as this organelle structure has
Drosophila homolog of the endosomal regulator His- been shown to not only downregulate surface receptors
domain protein tyrosine phosphatase (HD-PT) also but also to dynamically respond to intra- and extracellu-
known as PTPN23] was identified as a regulator of lar stimuli [171,172] and serve as an integrator for diverse
Yorkie endosomal localization [168]. A series of Hippo cellular signaling events [124,172].

Box 2. YAP and TAZ regulation in response to cellular stress


Cells are constantly subjected to external and internal stresses that stress, such as inhibition of glucose metabolism and ATP production,
endanger their integrity. YAP and TAZ are regulated in response to a induces AMP-activated protein kinase (AMPK)-mediated phosphoryla-
range of these cellular stresses (Figure I). Cytokinesis failure and extra tion of Angiomotin-like 1 (AMOTL1) to stabilize and increase AMOTL1,
centrosomes alter small G protein signaling and thereby activate which in turn stimulates LATS [35]. Furthermore, LATS also senses and
LATS2 kinase. LATS2 in turn inhibits YAP and TAZ and stabilizes p53, is activated by glucose starvation in an AMPK-independent manner
thus arresting cells at G1 phase to prevent tumorigenesis [143]. Treat- [201]. In addition, AMPK inhibits YAP by directly phosphorylating it at
ment of cancer cells with antitubulin drugs exerts a mitotic block, least two distinct sites [201,202]; therefore, energy stress inhibits YAP
which activates cyclin-dependent kinase 1 (CDK1) [194]. CDK1 directly nuclear activity by several mechanisms [35,201,202].
phosphorylates and inhibits YAP function to induce apoptosis.
However, the role of CDK1-mediated YAP phosphorylation is
controversial as mitotic phosphorylation of YAP by CDK1 promotes
mitotic defects and potentiates oncogenic functions of YAP
Energy stress
[195]. DNA damage activates the tyrosine kinase ABL through ATM–
re
JNK signaling, which in turn phosphorylates YAP to stimulate ilu Hy
fa po
proapoptotic functions of YAP in complex with p73 [196]. Endoplasmic is
es xia
reticulum (ER) stress has also recently been implicated in YAP okin AMPK
t
regulation. The ER stress transducer PKR-like ER kinase (PERK) Cy SIA
O H2
phosphorylates eukaryotic initiation factor 2a (eIF2a) during the RH LATS
adaptive stage of the unfolded protein response (UPR), which LA
TS TS
le arrest

Oxida

suppresses general protein synthesis and specifically induces the LA


M arrreest
s

translation of ATF4, which regulates YAP transcription [197]. ER stress


MST

ve stress
Cell cyc

thus induces YAP expression through the PERK–eIF2a–ATF4 axis to


LAT S
?
CDK1
G2//M

prevent cell death during the adaptive stage of the UPR, while
YAP/TAZ
prolonged ER stress activates Hippo signaling to inhibit YAP and
promote apoptosis [197].
L
AB

YAP and TAZ likewise respond to external stresses evoked by these


JNK

RH

cellular microenvironments. Hyperosmotic stress induces tyrosine


M

Sh chan
O
ge

Me
AT

4
e

ATF
ma

phosphorylation of TAZ by the ABL kinase, which facilitates the


ar

ABL
da

str al/

interaction between TAZ and nuclear factor of activated T cells 5 eIF2


α
ess
A

ic
DN

(NFAT5) to inhibit NFAT5 function in osmoregulatory transcription


K
[198]. YAP/TAZ are activated by interstitial flow-driven shear stress and PER
promote osteogenic differentiation of mesenchymal stem cells Osm
[147,199]. Rho GTPase appears to be involved in this regulation, s
tres stre
oc
although the precise mechanism remains unknown [148]. Oxidative ER s ss
stress evoked by ischemia and reperfusion in the mouse heart activates
the Hippo pathway to antagonize a functional YAP–FOXO1 complex,
TRENDS in Cell Biology
leading to enhanced oxidative stress-induced cell death [200]. By
contrast, hypoxia deactivates the Hippo pathway by destabilizing Figure I. Yes-associated protein (YAP) and transcriptional coactivator with a
LATS2 through SIAH2 ubiquitin ligase-induced degradation [33]. Energy PDZ-binding domain (TAZ) regulation in response to cellular stress.

508
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

Box 3. Outstanding questions References


1 Pan, D. (2010) The hippo signaling pathway in development and
 How is LATS activated in different contexts? cancer. Dev. Cell 19, 491–505
 How does the temporal and spatial organization and regulation of 2 Oh, H. and Irvine, K.D. (2010) Yorkie: the final destination of Hippo
the Hippo pathway occur? signaling. Trends Cell Biol. 20, 410–417
 Does the Hippo pathway regulate additional effectors besides YAP 3 Varelas, X. (2014) The Hippo pathway effectors TAZ and YAP in
and TAZ? development, homeostasis and disease. Development 141, 1614–1626
 How do YAP and TAZ differ and how are their nuclear/cytoplasmic 4 Moroishi, T. et al. (2015) The emerging roles of YAP and TAZ in
functions regulated? cancer. Nat. Rev. Cancer 15, 73–79
 What underlies the context-dependent physiological roles and 5 Harvey, K.F. et al. (2013) The Hippo pathway and human cancer. Nat.
activities of YAP and TAZ? Rev. Cancer 13, 246–257
 How can the Hippo pathway be therapeutically utilized in 6 Johnson, R. and Halder, G. (2014) The two faces of Hippo: targeting
regeneration and cancer? the Hippo pathway for regenerative medicine and cancer treatment.
Nat Rev. Drug Discov. 13, 63–79
7 Yu, F.X. et al. (2012) Regulation of the Hippo–YAP pathway by
With the realization that multiple signaling pathways G-protein-coupled receptor signaling. Cell 150, 780–791
converge and integrate the Hippo pathway, it will be impor- 8 Mo, J.S. et al. (2012) Regulation of the Hippo–YAP pathway by
tant to further delineate the interactions and dynamic protease-activated receptors (PARs). Genes Dev. 26, 2138–2143
9 Miller, E. et al. (2012) Identification of serum-derived sphingosine-
spatiotemporal reciprocal regulation of the Hippo pathway 1-phosphate as a small molecule regulator of YAP. Chem. Biol. 19,
to and from cellular signaling hubs such as the primary cilia 955–962
and the endolysosomal system to fully understand the 10 Bao, Y. et al. (2011) A cell-based assay to screen stimulators of the
contextual signaling mediated by YAP and TAZ and the Hippo pathway reveals the inhibitory effect of dobutamine on the
YAP-dependent gene transcription. J. Biochem. 150, 199–208
complete Hippo pathway.
11 Liu, G. et al. (2014) Kaposi sarcoma-associated herpesvirus promotes
tumorigenesis by modulating the Hippo pathway. Oncogene
Concluding remarks Published online September 8, 2014. http://dx.doi.org/10.1038/
The Hippo pathway extends beyond a simple kinase cas- onc.2014.281
cade targeting the transcription module and several pro- 12 Yu, F.X. et al. (2014) Mutant Gq/11 promote uveal melanoma
teins have been discovered to either completely or partially tumorigenesis by activating YAP. Cancer Cell 25, 822–830
bypass the core kinase module to regulate YAP and TAZ. 13 Feng, X. et al. (2014) Hippo-independent activation of YAP by the
GNAQ uveal melanoma oncogene through a trio-regulated rho
Studies in both Drosophila and mammals have broadened GTPase signaling circuitry. Cancer Cell 25, 831–845
the Hippo pathway network, which now extends to com- 14 Azzolin, L. et al. (2012) Role of TAZ as mediator of Wnt signaling. Cell
ponents of cell–cell junction protein complexes, additional 151, 1443–1456
kinases, cytoskeletal regulators, cellular organelles, and, 15 Varelas, X. et al. (2010) The Hippo pathway regulates Wnt/b-catenin
signaling. Dev. Cell 18, 579–591
most recently, cellular stress responses (Box 2). Numerous
16 Barry, E.R. et al. (2013) Restriction of intestinal stem cell expansion
pathways converge on multiple levels with YAP/TAZ and it and the regenerative response by YAP. Nature 493, 106–110
is being revealed that YAP/TAZ serve as potent manifolds 17 Konsavage, W.M., Jr et al. (2012) Wnt/b-catenin signaling regulates
by partnering with seemingly distant signaling molecules Yes-associated protein (YAP) gene expression in colorectal carcinoma
for contextual cellular decision making. cells. J. Biol. Chem. 287, 11730–11739
18 Hiemer, S.E. et al. (2014) The transcriptional regulators TAZ and YAP
Some of the initial in vivo Hippo pathway studies in flies
direct transforming growth factor beta-induced tumorigenic
[173] and mammals [27,174] highlighted the importance of phenotypes in breast cancer cells. J. Biol. Chem. 289, 13461–13474
this pathway in organ growth control as well as its thera- 19 Varelas, X. et al. (2008) TAZ controls Smad nucleocytoplasmic
peutic potential in regeneration and in tumorigenesis. shuttling and regulates human embryonic stem-cell self-renewal.
These studies laid the foundation for the immense interest Nat. Cell Biol. 10, 837–848
20 Serrano, I. et al. (2013) Inactivation of the Hippo tumour suppressor
in and research into this pathway. Many important ques- pathway by integrin-linked kinase. Nat. Commun. 4, 2976
tions remain to be answered (Box 3). Recent findings show- 21 Zhang, J. et al. (2009) YAP-dependent induction of amphiregulin
ing that increasing YAP activity in various organs can identifies a non-cell-autonomous component of the Hippo pathway.
reprogram fully differentiated cells into an undifferentiated Nat. Cell Biol. 11, 1444–1450
state in vivo [77,109,129] clearly indicate a powerful thera- 22 Fan, R. et al. (2013) Regulation of Hippo pathway by mitogenic
growth factors via phosphoinositide 3-kinase and phosphoinositide-
peutic capacity in tissue regeneration and repair. Inhibiting dependent kinase-1. Proc. Natl. Acad. Sci. U.S.A. 110, 2569–2574
the upstream Hippo kinase module should be a prime target 23 Reddy, B.V. and Irvine, K.D. (2013) Regulation of Hippo signaling by
in regenerative medicine [100] and research in cellular EGFR–MAPK signaling through Ajuba family proteins. Dev. Cell 24,
plasticity should be explored in the future. A clear challenge 459–471
24 Haskins, J.W. et al. (2014) Neuregulin 1-activated ERBB4 interacts
for this therapeutic strategy will be to fine-tune the activa-
with YAP to induce Hippo pathway target genes and promote cell
tion state of YAP/TAZ, recollecting the interdependence migration. Sci. Signal. 7, ra116
between cancer and regeneration [4,6,100]. 25 Li, Y. et al. (2012) Genome-wide analysis of N1ICD/RBPJ targets in
vivo reveals direct transcriptional regulation of Wnt, SHH, and Hippo
Acknowledgments pathway effectors by Notch1. Stem Cells 30, 741–752
The authors apologize to colleagues whose work could not be cited owing to 26 Tschaharganeh, D.F. et al. (2013) Yes-associated protein up-regulates
space limitations. The authors thank all members of the Guan laboratory Jagged-1 and activates the Notch pathway in human hepatocellular
for insightful discussion and critical comments. Work in the Guan carcinoma. Gastroenterology 144, 1530–1542
laboratory was supported by a National Institutes of Health (NIH) grant 27 Camargo, F.D. et al. (2007) YAP1 increases organ size and expands
(CA132809 and EYO226116) to K-L.G. C.G.H. is supported by a undifferentiated progenitor cells. Curr. Biol. 17, 2054–2060
Postdoctoral Fellowship from the Danish Council for Independent Re- 28 Rayon, T. et al. (2014) Notch and Hippo converge on cdx2 to specify
searchjNatural Sciences. T.M. is supported by the Japan Society for the the trophectoderm lineage in the mouse blastocyst. Dev. Cell 30,
Promotion of Science (JSPS) Postdoctoral Fellowships for Research Abroad. 410–422

509
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

29 Hergovich, A. (2013) Regulation and functions of mammalian LATS/ 57 Liu, X. et al. (2013) PTPN14 interacts with and negatively regulates
NDR kinases: looking beyond canonical Hippo signalling. Cell Biosci. the oncogenic function of YAP. Oncogene 32, 1266–1273
3, 32 58 Wang, W. et al. (2012) PTPN14 is required for the density-dependent
30 Zhou, D. et al. (2009) Mst1 and Mst2 maintain hepatocyte quiescence control of YAP1. Genes Dev. 26, 1959–1971
and suppress hepatocellular carcinoma development through 59 Schlegelmilch, K. et al. (2011) Yap1 acts downstream of a-catenin to
inactivation of the Yap1 oncogene. Cancer Cell 16, 425–438 control epidermal proliferation. Cell 144, 782–795
31 Yin, F. et al. (2013) Spatial organization of Hippo signaling at the 60 Sudol, M. and Harvey, K.F. (2010) Modularity in the Hippo signaling
plasma membrane mediated by the tumor suppressor Merlin/NF2. pathway. Trends Biochem. Sci. 35, 627–633
Cell 154, 1342–1355 61 Mana-Capelli, S. et al. (2014) Angiomotins link F-actin architecture to
32 Li, Q. et al. (2014) The conserved Misshapen–Warts–Yorkie pathway Hippo pathway signaling. Mol. Biol. Cell 25, 1676–1685
acts in enteroblasts to regulate intestinal stem cells in Drosophila. 62 Adler, J.J. et al. (2013) Serum deprivation inhibits the transcriptional
Dev. Cell 31, 291–304 co-activator YAP and cell growth via phosphorylation of the 130-kDa
33 Ma, B. et al. (2015) Hypoxia regulates Hippo signalling through the isoform of Angiomotin by the LATS1/2 protein kinases. Proc. Natl.
SIAH2 ubiquitin E3 ligase. Nat. Cell Biol. 17, 95–103 Acad. Sci. U.S.A. 110, 17368–17373
34 Lignitto, L. et al. (2013) Proteolysis of MOB1 by the ubiquitin ligase 63 Dai, X. et al. (2013) Phosphorylation of Angiomotin by Lats1/2 kinases
praja2 attenuates Hippo signalling and supports glioblastoma inhibits F-actin binding, cell migration, and angiogenesis. J. Biol.
growth. Nat. Commun. 4, 1822 Chem. 288, 34041–34051
35 DeRan, M. et al. (2014) Energy stress regulates Hippo–YAP signaling 64 Yi, C. et al. (2013) The p130 isoform of Angiomotin is required for Yap-
involving AMPK-mediated regulation of Angiomotin-like 1 protein. mediated hepatic epithelial cell proliferation and tumorigenesis. Sci.
Cell Rep. 9, 495–503 Signal. 6, ra77
36 Salah, Z. et al. (2011) Negative regulation of the Hippo pathway by E3 65 Chan, S.W. et al. (2011) WW domain-mediated interaction with
ubiquitin ligase ITCH is sufficient to promote tumorigenicity. Cancer Wbp2 is important for the oncogenic property of TAZ. Oncogene
Res. 71, 2010–2020 30, 600–610
37 Bae, S.J. et al. (2015) NEDD4 controls intestinal stem cell 66 Zhang, X. et al. (2011) Wbp2 cooperates with Yorkie to drive tissue
homeostasis by regulating the Hippo signalling pathway. Nat. growth downstream of the Salvador–Warts–Hippo pathway. Cell
Commun. 6, 6314 Death Differ. 18, 1346–1355
38 Heidary Arash, E. et al. (2014) Arhgef7 promotes activation of the 67 Sansores-Garcia, L. et al. (2013) Mask is required for the activity of the
Hippo pathway core kinase Lats. EMBO J. 33, 2997–3011 Hippo pathway effector Yki/YAP. Curr. Biol. 23, 229–235
39 Pearson, H.B. et al. (2011) SCRIB expression is deregulated in human 68 Sidor, C.M. et al. (2013) Mask proteins are cofactors of Yorkie/YAP in
prostate cancer, and its deficiency in mice promotes prostate the Hippo pathway. Curr. Biol. 23, 223–228
neoplasia. J. Clin. Invest. 121, 4257–4267 69 Koontz, L.M. et al. (2013) The Hippo effector Yorkie controls normal
40 Liu-Chittenden, Y. et al. (2012) Genetic and pharmacological tissue growth by antagonizing Scalloped-mediated default repression.
disruption of the TEAD–YAP complex suppresses the oncogenic Dev. Cell 25, 388–401
activity of YAP. Genes Dev. 26, 1300–1305 70 Zhang, W. et al. (2014) VGLL4 functions as a new tumor suppressor in
41 Cockburn, K. et al. (2013) The Hippo pathway member Nf2 is required lung cancer by negatively regulating the YAP–TEAD transcriptional
for inner cell mass specification. Curr. Biol. 23, 1195–1201 complex. Cell Res. 24, 331–343
42 Zhao, B. et al. (2007) Inactivation of YAP oncoprotein by the Hippo 71 Jiao, S. et al. (2014) A peptide mimicking VGLL4 function acts as
pathway is involved in cell contact inhibition and tissue growth a YAP antagonist therapy against gastric cancer. Cancer Cell 25,
control. Genes Dev. 21, 2747–2761 166–180
43 Zhao, B. et al. (2008) TEAD mediates YAP-dependent gene induction 72 Vassilev, A. et al. (2001) TEAD/TEF transcription factors utilize the
and growth control. Genes Dev. 22, 1962–1971 activation domain of YAP65, a Src/Yes-associated protein localized in
44 Kim, N.G. et al. (2011) E-cadherin mediates contact inhibition of the cytoplasm. Genes Dev. 15, 1229–1241
proliferation through Hippo signaling-pathway components. Proc. 73 Ota, M. and Sasaki, H. (2008) Mammalian Tead proteins regulate cell
Natl. Acad. Sci. U.S.A. 108, 11930–11935 proliferation and contact inhibition as transcriptional mediators of
45 Cordenonsi, M. et al. (2011) The Hippo transducer TAZ confers cancer Hippo signaling. Development 135, 4059–4069
stem cell-related traits on breast cancer cells. Cell 147, 759–772 74 Mo, J.S. et al. (2014) The Hippo signaling pathway in stem cell biology
46 Baumgartner, R. et al. (2010) The WW domain protein Kibra acts and cancer. EMBO Rep. 15, 642–656
upstream of Hippo in Drosophila. Dev. Cell 18, 309–316 75 Oh, H. et al. (2014) Yorkie promotes transcription by recruiting a
47 Genevet, A. et al. (2010) Kibra is a regulator of the Salvador/Warts/ histone methyltransferase complex. Cell Rep. 8, 449–459
Hippo signaling network. Dev. Cell 18, 300–308 76 Qing, Y. et al. (2014) The Hippo effector Yorkie activates transcription
48 Yu, J. et al. (2010) Kibra functions as a tumor suppressor protein that by interacting with a histone methyltransferase complex through
regulates Hippo signaling in conjunction with Merlin and Expanded. Ncoa6. Elife 3, e02564
Dev. Cell 18, 288–299 77 Skibinski, A. et al. (2014) The Hippo transducer TAZ interacts with
49 Hirate, Y. et al. (2013) Polarity-dependent distribution of Angiomotin the SWI/SNF complex to regulate breast epithelial lineage
localizes Hippo signaling in preimplantation embryos. Curr. Biol. 23, commitment. Cell Rep. 6, 1059–1072
1181–1194 78 Oh, H. et al. (2013) Genome-wide association of Yorkie with chromatin
50 Li, W. et al. (2014) Merlin/NF2 loss-driven tumorigenesis linked to and chromatin-remodeling complexes. Cell Rep. 3, 309–318
CRL4DCAF1-mediated inhibition of the hippo pathway kinases Lats1 79 Beyer, T.A. et al. (2013) Switch enhancers interpret TGF-b and Hippo
and 2 in the nucleus. Cancer Cell 26, 48–60 signaling to control cell fate in human embryonic stem cells. Cell Rep.
51 Das Thakur, M. et al. (2010) Ajuba LIM proteins are negative 5, 1611–1624
regulators of the Hippo signaling pathway. Curr. Biol. 20, 657–662 80 Kim, M. et al. (2015) Transcriptional co-repressor function of the
52 Rauskolb, C. et al. (2014) Cytoskeletal tension inhibits Hippo Hippo pathway transducers YAP and TAZ. Cell Rep. 11, 270–282
signaling through an Ajuba–Warts complex. Cell 158, 143–156 81 Mori, M. et al. (2014) Hippo signaling regulates microprocessor and
53 Zhao, B. et al. (2011) Angiomotin is a novel Hippo pathway component links cell-density-dependent miRNA biogenesis to cancer. Cell 156,
that inhibits YAP oncoprotein. Genes Dev. 25, 51–63 893–906
54 Chan, S.W. et al. (2011) Hippo pathway-independent restriction of 82 Chaulk, S.G. et al. (2014) The Hippo pathway effectors TAZ/YAP
TAZ and YAP by Angiomotin. J. Biol. Chem. 286, 7018–7026 regulate Dicer expression and microRNA biogenesis through Let-7.
55 Wang, W. et al. (2011) Angiomotin-like proteins associate with and J. Biol. Chem. 289, 1886–1891
negatively regulate YAP1. J. Biol. Chem. 286, 4364–4370 83 Tumaneng, K. et al. (2012) YAP mediates crosstalk between the Hippo
56 Leung, C.Y. and Zernicka-Goetz, M. (2013) Angiomotin prevents and PI3K–TOR pathways by suppressing PTEN via miR-29. Nat. Cell
pluripotent lineage differentiation in mouse embryos via Hippo Biol. 14, 1322–1329
pathway-dependent and -independent mechanisms. Nat. Commun. 84 Piccolo, S. et al. (2014) The biology of YAP/TAZ: Hippo signaling and
4, 2251 beyond. Physiol. Rev. 94, 1287–1312

510
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

85 Clevers, H. et al. (2014) Stem cell signaling. An integral program for 111 Judson, R.N. et al. (2012) The Hippo pathway member Yap plays a key
tissue renewal and regeneration: Wnt signaling and stem cell control. role in influencing fate decisions in muscle satellite cells. J. Cell Sci.
Science 346, 1248012 125, 6009–6019
86 Imajo, M. et al. (2012) A molecular mechanism that links Hippo 112 Lee, M.J. et al. (2014) YAP and TAZ regulate skin wound healing.
signalling to the inhibition of Wnt/b-catenin signalling. EMBO J. J. Invest. Dermatol. 134, 518–525
31, 1109–1122 113 Diepenbruck, M. et al. (2014) Tead2 expression levels control the
87 Azzolin, L. et al. (2014) YAP/TAZ incorporation in the b-catenin subcellular distribution of Yap and Taz, zyxin expression and
destruction complex orchestrates the Wnt response. Cell 158, 157–170 epithelial–mesenchymal transition. J. Cell Sci. 127, 1523–1536
88 Takahashi, A. et al. (2011) SHP2 tyrosine phosphatase converts 114 Kim, M. et al. (2013) cAMP/PKA signalling reinforces the LATS–YAP
parafibromin/Cdc73 from a tumor suppressor to an oncogenic pathway to fully suppress YAP in response to actin cytoskeletal
driver. Mol. Cell 43, 45–56 changes. EMBO J. 32, 1543–1555
89 Tsutsumi, R. et al. (2013) YAP and TAZ, Hippo signaling targets, act 115 Yu, F.X. et al. (2013) Protein kinase A activates the Hippo pathway to
as a rheostat for nuclear SHP2 function. Dev. Cell 26, 658–665 modulate cell proliferation and differentiation. Genes Dev. 27, 1223–
90 Mosimann, C. et al. (2006) Parafibromin/Hyrax activates Wnt/Wg 1232
target gene transcription by direct association with b-catenin/ 116 Plouffe, S.W. et al. (2015) Disease implications of the Hippo/YAP
Armadillo. Cell 125, 327–341 pathway. Trends Mol. Med. 21, 212–222
91 Llado, V. et al. (2015) Repression of intestinal stem cell function and 117 Park, H.W. and Guan, K.L. (2013) Regulation of the Hippo pathway
tumorigenesis through direct phosphorylation of b-catenin and Yap and implications for anticancer drug development. Trends
by PKCz. Cell Rep. 10, 740–754 Pharmacol. Sci. 34, 581–589
92 Heallen, T. et al. (2011) Hippo pathway inhibits Wnt signaling to 118 Yun, C.C. and Kumar, A. (2014) Diverse roles of LPA signaling in the
restrain cardiomyocyte proliferation and heart size. Science 332, intestinal epithelium. Exp. Cell Res. 333, 201–207
458–461 119 Fukui, H. et al. (2014) S1P–Yap1 signaling regulates endoderm
93 Cai, J. et al. (2010) The Hippo signaling pathway restricts the formation required for cardiac precursor cell migration in
oncogenic potential of an intestinal regeneration program. Genes zebrafish. Dev. Cell 31, 128–136
Dev. 24, 2383–2388 120 Avery, K. et al. (2008) Sphingosine-1-phosphate mediates
94 Hossain, Z. et al. (2007) Glomerulocystic kidney disease in mice with a transcriptional regulation of key targets associated with survival,
targeted inactivation of Wwtr1. Proc. Natl. Acad. Sci. U.S.A. 104, proliferation, and pluripotency in human embryonic stem cells. Stem
1631–1636 Cells Dev. 17, 1195–1205
95 Zhou, D. et al. (2011) Mst1 and Mst2 protein kinases restrain 121 Nagata, Y. et al. (2006) Entry of muscle satellite cells into the cell cycle
intestinal stem cell proliferation and colonic tumorigenesis by requires sphingolipid signaling. J. Cell Biol. 174, 245–253
inhibition of Yes-associated protein (Yap) overabundance. Proc. 122 Komuro, A. et al. (2003) WW domain-containing protein YAP
Natl. Acad. Sci. U.S.A. 108, E1312–E1320 associates with ErbB-4 and acts as a co-transcriptional activator
96 Imajo, M. et al. (2015) Dual role of YAP and TAZ in renewal of the for the carboxyl-terminal fragment of ErbB-4 that translocates to
intestinal epithelium. Nat. Cell Biol. 17, 7–19 the nucleus. J. Biol. Chem. 278, 33334–33341
97 Lai, D. and Yang, X. (2013) BMP4 is a novel transcriptional target and 123 Omerovic, J. et al. (2004) Ligand-regulated association of ErbB-4 to
mediator of mammary cell migration downstream of the Hippo the transcriptional co-activator YAP65 controls transcription at the
pathway component TAZ. Cell. Signal. 25, 1720–1728 nuclear level. Exp. Cell Res. 294, 469–479
98 Seo, E. et al. (2013) SOX2 regulates YAP1 to maintain stemness 124 Tomas, A. et al. (2014) EGF receptor trafficking: consequences for
and determine cell fate in the osteo–adipo lineage. Cell Rep. 3, 2075– signaling and cancer. Trends Cell Biol. 24, 26–34
2087 125 Nishioka, N. et al. (2009) The Hippo signaling pathway components
99 Haramis, A.P. et al. (2004) De novo crypt formation and juvenile Lats and Yap pattern Tead4 activity to distinguish mouse
polyposis on BMP inhibition in mouse intestine. Science 303, 1684– trophectoderm from inner cell mass. Dev. Cell 16, 398–410
1686 126 Home, P. et al. (2012) Altered subcellular localization of transcription
100 Forbes, S.J. and Rosenthal, N. (2014) Preparing the ground for tissue factor TEAD4 regulates first mammalian cell lineage commitment.
regeneration: from mechanism to therapy. Nat. Med. 20, 857–869 Proc. Natl. Acad. Sci. U.S.A. 109, 7362–7367
101 Taniguchi, K. et al. (2015) A gp130–Src–YAP module links 127 Yagi, R. et al. (2007) Transcription factor TEAD4 specifies the
inflammation to epithelial regeneration. Nature 519, 57–62 trophectoderm lineage at the beginning of mammalian development.
102 Massague, J. (2012) TGFb signalling in context. Nat. Rev. Mol. Cell Development 134, 3827–3836
Biol. 13, 616–630 128 Strumpf, D. et al. (2005) Cdx2 is required for correct cell fate
103 Schuldiner, M. et al. (2000) Effects of eight growth factors on the specification and differentiation of trophectoderm in the mouse
differentiation of cells derived from human embryonic stem cells. blastocyst. Development 132, 2093–2102
Proc. Natl. Acad. Sci. U.S.A. 97, 11307–11312 129 Yimlamai, D. et al. (2014) Hippo pathway activity influences liver cell
104 Varelas, X. et al. (2010) The Crumbs complex couples cell density fate. Cell 157, 1324–1338
sensing to Hippo-dependent control of the TGF-b–SMAD pathway. 130 Tchorz, J.S. et al. (2009) Notch2 signaling promotes biliary epithelial
Dev. Cell 19, 831–844 cell fate specification and tubulogenesis during bile duct development
105 Nallet-Staub, F. et al. (2015) Cell density sensing alters TGF-b in mice. Hepatology 50, 871–879
signaling in a cell-type-specific manner, independent from Hippo 131 Boulter, L. et al. (2012) Macrophage-derived Wnt opposes Notch
pathway activation. Dev. Cell 32, 640–651 signaling to specify hepatic progenitor cell fate in chronic liver
106 Narimatsu, M. et al. (2015) Distinct polarity cues direct Taz/Yap and disease. Nat. Med. 18, 572–579
TGFb receptor localization to differentially control TGFb-induced 132 Bjornson, C.R. et al. (2012) Notch signaling is necessary to
Smad signaling. Dev. Cell 32, 652–656 maintain quiescence in adult muscle stem cells. Stem Cells 30,
107 Lian, I. et al. (2010) The role of YAP transcription coactivator in 232–242
regulating stem cell self-renewal and differentiation. Genes Dev. 24, 133 Tremblay, A.M. et al. (2014) The Hippo transducer YAP1 transforms
1106–1118 activated satellite cells and is a potent effector of embryonal
108 Alarcon, C. et al. (2009) Nuclear CDKs drive Smad transcriptional rhabdomyosarcoma formation. Cancer Cell 26, 273–287
activation and turnover in BMP and TGF-b pathways. Cell 139, 757– 134 Watt, K.I. et al. (2010) Yap is a novel regulator of C2C12 myogenesis.
769 Biochem. Biophys. Res. Commun. 393, 619–624
109 Zhao, R. et al. (2014) Yap tunes airway epithelial size and architecture 135 Wang, N. et al. (2009) Mechanotransduction at a distance:
by regulating the identity, maintenance, and self-renewal of stem mechanically coupling the extracellular matrix with the nucleus.
cells. Dev. Cell 30, 151–165 Nat. Rev. Mol. Cell Biol. 10, 75–82
110 Mahoney, J.E. et al. (2014) The Hippo pathway effector Yap controls 136 Seguin, L. et al. (2015) Integrins and cancer: regulators of cancer
patterning and differentiation of airway epithelial progenitors. Dev. stemness, metastasis, and drug resistance. Trends Cell Biol. 25,
Cell 30, 137–150 234–240

511
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

137 Gaspar, P. and Tapon, N. (2014) Sensing the local environment: actin 166 Wang, W. et al. (2014) Defining the protein–protein interaction network
architecture and Hippo signalling. Curr. Opin. Cell Biol. 31, 74–83 of the human Hippo pathway. Mol. Cell. Proteomics 13, 119–131
138 Engler, A.J. et al. (2006) Matrix elasticity directs stem cell lineage 167 Couzens, A.L. et al. (2013) Protein interaction network of the
specification. Cell 126, 677–689 mammalian Hippo pathway reveals mechanisms of kinase–
139 Zhao, B. et al. (2012) Cell detachment activates the Hippo pathway phosphatase interactions. Sci. Signal. 6, rs15
via cytoskeleton reorganization to induce anoikis. Genes Dev. 26, 168 Gilbert, M.M. et al. (2011) A screen for conditional growth suppressor
54–68 genes identifies the Drosophila homolog of HD-PTP as a regulator of
140 Cui, Y. et al. (2015) Cyclic stretching of soft substrates induces the oncoprotein Yorkie. Dev. Cell 20, 700–712
spreading and growth. Nat. Commun. 6, 6333 169 Liang, N. et al. (2014) Regulation of YAP by mTOR and autophagy
141 Wada, K. et al. (2011) Hippo pathway regulation by cell morphology reveals a therapeutic target of tuberous sclerosis complex. J. Exp.
and stress fibers. Development 138, 3907–3914 Med. 211, 2249–2263
142 Dupont, S. et al. (2011) Role of YAP/TAZ in mechanotransduction. 170 Zhao, B. et al. (2010) A coordinated phosphorylation by Lats and CK1
Nature 474, 179–183 regulates YAP stability through SCFb-TRCP. Genes Dev. 24, 72–85
143 Ganem, N.J. et al. (2014) Cytokinesis failure triggers Hippo tumor 171 Kim, Y.C. and Guan, K.L. (2015) mTOR: a pharmacologic target for
suppressor pathway activation. Cell 158, 833–848 autophagy regulation. J. Clin. Invest. 125, 25–32
144 Fletcher, G.C. et al. (2015) The Spectrin cytoskeleton regulates the 172 Jewell, J.L. et al. (2013) Amino acid signalling upstream of mTOR.
Hippo signalling pathway. EMBO J. 34, 940–954 Nat. Rev. Mol. Cell Biol. 14, 133–139
145 Deng, H. et al. (2015) Spectrin regulates Hippo signaling by 173 Justice, R.W. et al. (1995) The Drosophila tumor suppressor gene warts
modulating cortical actomyosin activity. Elife 4, e06567 encodes a homolog of human myotonic dystrophy kinase and is required
146 Chang, C. et al. (2015) A laminin 511 matrix is regulated by TAZ and for the control of cell shape and proliferation. Genes Dev. 9, 534–546
functions as the ligand for the a6Bb1 integrin to sustain breast cancer 174 Dong, J. et al. (2007) Elucidation of a universal size-control
stem cells. Genes Dev. 29, 1–6 mechanism in Drosophila and mammals. Cell 130, 1120–1133
147 Kim, K.M. et al. (2014) Shear stress induced by an interstitial level of 175 Hilman, D. and Gat, U. (2011) The evolutionary history of YAP and
slow flow increases the osteogenic differentiation of mesenchymal the Hippo/YAP pathway. Mol. Biol. Evol. 28, 2403–2417
stem cells through TAZ activation. PLoS ONE 9, e92427 176 Sebe-Pedros, A. et al. (2012) Premetazoan origin of the Hippo
148 Halder, G. et al. (2012) Transduction of mechanical and cytoskeletal signaling pathway. Cell Rep. 1, 13–20
cues by YAP and TAZ. Nat. Rev. Mol. Cell Biol. 13, 591–600 177 Boggiano, J.C. et al. (2011) Tao-1 phosphorylates Hippo/MST kinases
149 Hong, J.H. et al. (2005) TAZ, a transcriptional modulator of to regulate the Hippo–Salvador–Warts tumor suppressor pathway.
mesenchymal stem cell differentiation. Science 309, 1074–1078 Dev. Cell 21, 888–895
150 Parsons, J.T. et al. (2010) Cell adhesion: integrating cytoskeletal 178 Poon, C.L. et al. (2011) The sterile 20-like kinase Tao-1 controls tissue
dynamics and cellular tension. Nat. Rev. Mol. Cell Biol. 11, growth by regulating the Salvador–Warts–Hippo pathway. Dev. Cell
633–643 21, 896–906
151 Wu, D.D. et al. (2014) Matricellular protein Cyr61 bridges 179 Huang, H.L. et al. (2013) Par-1 regulates tissue growth by influencing
lysophosphatidic acid and integrin pathways leading to cell Hippo phosphorylation status and Hippo–Salvador association. PLoS
migration. J. Biol. Chem. 289, 5774–5783 Biol. 11, e1001620
152 Tang, Y. et al. (2013) MT1-MMP-dependent control of skeletal stem 180 Wehr, M.C. et al. (2013) Salt-inducible kinases regulate growth
cell commitment via a b1-integrin/YAP/TAZ signaling axis. Dev. Cell through the Hippo signalling pathway in Drosophila. Nat. Cell
25, 402–416 Biol. 15, 61–71
153 Kim, M. et al. (2014) The MST1/2–SAV1 complex of the Hippo 181 Polesello, C. et al. (2006) The Drosophila RASSF homolog antagonizes
pathway promotes ciliogenesis. Nat. Commun. 5, 5370 the Hippo pathway. Curr. Biol. 16, 2459–2465
154 Kang, H.S. et al. (2009) Glis3 is associated with primary cilia and 182 Ribeiro, P.S. et al. (2010) Combined functional genomic and proteomic
Wwtr1/TAZ and implicated in polycystic kidney disease. Mol. Cell. approaches identify a PP2A complex as a negative regulator of Hippo
Biol. 29, 2556–2569 signaling. Mol. Cell 39, 521–534
155 Kim, J. et al. (2015) Actin remodelling factors control ciliogenesis by 183 Mohseni, M. et al. (2014) A genetic screen identifies an LKB1–MARK
regulating YAP/TAZ activity and vesicle trafficking. Nat. Commun. 6, signalling axis controlling the Hippo–YAP pathway. Nat. Cell Biol.
6781 16, 108–117
156 Czarnecki, P.G. and Shah, J.V. (2012) The ciliary transition zone: 184 Praskova, M. et al. (2004) Regulation of the MST1 kinase by
from morphology and molecules to medicine. Trends Cell Biol. 22, autophosphorylation, by the growth inhibitory proteins, RASSF1
201–210 and NORE1, and by Ras. Biochem. J. 381, 453–462
157 Battle, C. et al. (2015) Intracellular and extracellular forces drive 185 Ikeda, M. et al. (2009) Hippo pathway-dependent and -independent
primary cilia movement. Proc. Natl. Acad. Sci. U.S.A. 112, 1410–1415 roles of RASSF6. Sci. Signal. 2, ra59
158 Fernandez, L.A. et al. (2009) YAP1 is amplified and up-regulated in 186 Bossuyt, W. et al. (2014) An evolutionary shift in the regulation of the
hedgehog-associated medulloblastomas and mediates sonic Hippo pathway between mice and flies. Oncogene 33, 1218–1228
hedgehog-driven neural precursor proliferation. Genes Dev. 23, 187 Cappello, S. et al. (2013) Mutations in genes encoding the cadherin
2729–2741 receptor–ligand pair DCHS1 and FAT4 disrupt cerebral cortical
159 Sang, L. et al. (2011) Mapping the NPHP–JBTS–MKS protein development. Nat. Genet. 45, 1300–1308
network reveals ciliopathy disease genes and pathways. Cell 145, 188 Chen, C.L. et al. (2010) The apical–basal cell polarity determinant
513–528 Crumbs regulates Hippo signaling in Drosophila. Proc. Natl. Acad.
160 Habbig, S. et al. (2012) The ciliopathy disease protein NPHP9 Sci. U.S.A. 107, 15810–15815
promotes nuclear delivery and activation of the oncogenic 189 Ling, C. et al. (2010) The apical transmembrane protein Crumbs
transcriptional regulator TAZ. Hum. Mol. Genet. 21, 5528–5538 functions as a tumor suppressor that regulates Hippo signaling
161 Frank, V. et al. (2013) Mutations in NEK8 link multiple organ by binding to Expanded. Proc. Natl. Acad. Sci. U.S.A. 107, 10532–10537
dysplasia with altered Hippo signalling and increased c-MYC 190 Robinson, B.S. et al. (2010) Crumbs regulates Salvador/Warts/Hippo
expression. Hum. Mol. Genet. 22, 2177–2185 signaling in Drosophila via the FERM-domain protein Expanded.
162 Briscoe, J. and Therond, P.P. (2013) The mechanisms of hedgehog Curr. Biol. 20, 582–590
signalling and its roles in development and disease. Nat. Rev. Mol. 191 Badouel, C. et al. (2009) The FERM-domain protein Expanded
Cell Biol. 14, 416–429 regulates Hippo pathway activity via direct interactions with the
163 Sharpe, H.J. et al. (2015) Regulation of the oncoprotein Smoothened transcriptional activator Yorkie. Dev. Cell 16, 411–420
by small molecules. Nat. Chem. Biol. 11, 246–255 192 Oh, H. et al. (2009) Phosphorylation-independent repression of Yorkie
164 Hildebrandt, F. et al. (2011) Ciliopathies. N. Engl. J. Med. 364, 1533– in Fat–Hippo signaling. Dev. Biol. 335, 188–197
1543 193 Hamaratoglu, F. et al. (2006) The tumour-suppressor genes NF2/
165 Kwon, Y. et al. (2013) The Hippo signaling pathway interactome. Merlin and Expanded act through Hippo signalling to regulate cell
Science 342, 737–740 proliferation and apoptosis. Nat. Cell Biol. 8, 27–36

512
Feature Review Trends in Cell Biology September 2015, Vol. 25, No. 9

194 Zhao, Y. et al. (2014) YAP-induced resistance of cancer cells to 198 Jang, E.J. et al. (2012) TAZ suppresses NFAT5 activity through
antitubulin drugs is modulated by a Hippo-independent pathway. tyrosine phosphorylation. Mol. Cell. Biol. 32, 4925–4932
Cancer Res. 74, 4493–4503 199 Zhong, W. et al. (2013) Mesenchymal stem cell and chondrocyte fates
195 Yang, S. et al. (2013) CDK1 phosphorylation of YAP promotes mitotic in a multishear microdevice are regulated by Yes-associated protein.
defects and cell motility and is essential for neoplastic Stem Cells Dev. 22, 2083–2093
transformation. Cancer Res. 73, 6722–6733 200 Shao, D. et al. (2014) A functional interaction between Hippo–YAP
196 Basu, S. et al. (2003) Akt phosphorylates the Yes-associated protein, signalling and FoxO1 mediates the oxidative stress response. Nat.
YAP, to induce interaction with 14-3-3 and attenuation of p73- Commun. 5, 3315
mediated apoptosis. Mol. Cell 11, 11–23 201 Wang, W. et al. (2015) AMPK modulates Hippo pathway activity to
197 Wu, H. et al. (2015) Integration of Hippo signalling and the unfolded regulate energy homeostasis. Nat. Cell Biol. 17, 490–499
protein response to restrain liver overgrowth and tumorigenesis. Nat. 202 Mo, J.S. et al. (2015) Cellular energy stress induces AMPK-mediated
Commun. 6, 6239 regulation of YAP and the Hippo pathway. Nat. Cell Biol. 17, 500–510

513

You might also like