Sei sulla pagina 1di 28

BBA - Reviews on Cancer 1868 (2017) 456–483

Contents lists available at ScienceDirect

BBA - Reviews on Cancer


journal homepage: www.elsevier.com/locate/bbacan

Review

The roles of ubiquitin modifying enzymes in neoplastic disease MARK


a,b a a,b,c a
Nishi Kumari , Patrick William Jaynes , Azad Saei , Prasanna Vasudevan Iyengar ,
John Lalith Charles Richarda, Pieter Johan Adam Eichhorna,b,⁎
a
Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
b
Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
c
Genome Institute of Singapore, A*STAR, Singapore

A R T I C L E I N F O A B S T R A C T

Keywords: The initial experiments performed by Rose, Hershko, and Ciechanover describing the identification of a specific
Ubiquitin degradation signal in short-lived proteins paved the way to the discovery of the ubiquitin mediated regulation of
E3 ligases numerous physiological functions required for cellular homeostasis. Since their discovery of ubiquitin and
Deubiquitinating enzymes ubiquitin function over 30 years ago it has become wholly apparent that ubiquitin and their respective ubiquitin
Cancer
modifying enzymes are key players in tumorigenesis. The human genome encodes approximately 600 putative
Dub inhibitors
E3 ligases and 80 deubiquitinating enzymes and in the majority of cases these enzymes exhibit specificity in
sustaining either pro-tumorigenic or tumour repressive responses. In this review, we highlight the known on-
cogenic and tumour suppressive effects of ubiquitin modifying enzymes in cancer relevant pathways with spe-
cific focus on PI3K, MAPK, TGFβ, WNT, and YAP pathways. Moreover, we discuss the capacity of targeting DUBs
as a novel anticancer therapeutic strategy.

1. Introduction enzymes (DUBs) as tumour suppressors or oncogenes has also been an


area of intense research. In addition to E3 ligase function here we
The initial concept of protein turnover was viewed as a minor provide a portrait of DUB mutational profiles and their respective
component in cellular homeostasis for it was thought that proteins in substrate specificity in contribution to tumorigenesis.
general were intrinsically stable and were only subjected to degradation The ubiquitin moiety is a highly conserved 76 amino acid poly-
when proteins were damage by prolonged “wear and tear”. It is of peptide and ubiquitination of target proteins by ubiquitin and ubiquitin
course now recognized that ubiquitination mediated proteosomal de- like molecules serves as mechanism to regulate a myriad of protein
gradation of proteins is far more wide-spread and dynamically regu- functions including protein stability, subcellular localisation, and ac-
lated then was originally imagined. Furthermore, developments over tivity. The ubiquitination cascade comprises of coordinated action of a
the last two decades has led to a better understanding of the versatile E1 activating enzyme, a E2 ubiquitin-conjugating enzyme, and the E3
functions of ubiquitination. We now understand that ubiquitin is able to ubiquitin protein ligase. E1 ligases act as ubiquitin sponges recruiting
form eight structurally and functionally distinct polymers which can free ubiquitin inside the cell and subsequently transferring ubiquitin to
affect not only protein stability but also affect protein activity and the E2 ligase. The major component of the ubiquitin cascade is the E3
cellular localization. Accumulating evidence indicates that perturba- ligases of which the human genome encodes over 600 allowing a high
tions affecting the activity of ubiquitin modifying enzymes are im- degree of substrate specificity. Three main families of E3 ligases exist:
portant in tumour cell growth and survival and therefore these enzymes homologues to E6-associated protein carboxy terminus (HECT) family,
have been suggested to be potential targets for pharmacological inter- really interesting new gene (RING) family, which are the most abun-
vention. Clinical validation of this was initially observed with the ap- dant in the genome, and RING-Between-RING (RBR) family. All three
proval of the proteasome inhibitor Bortezomib in multiple myeloma families link E2s with substrates however, ubiquitin transfer is in-
with patients exhibiting significant responses to the treatment [1]. The herently unique between the E3s. In the case of HECT domain ligases
involvement of E3 ligases in crucial signalling pathways implicated in ubiquitin transfer involves the formation of a thioester intermediate
tumour progression is well documented and a number of excellent re- formed between ubiquitin and the catalytic cysteine of the HECT E3
views have been written on the role of E3 ligases in cancer which we prior to transfer of ubiquitin to its recruited substrate [2]. Unlike the
invite the reviewer to read [2–5]. However, the role of deubiquitinating HECT domain ligases, RING ligases appear to act as scaffolds permitting


Corresponding author at: Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive 12-01, 117599, Singapore.
E-mail address: pieter_eichhorn@nus.edu.sg (P.J.A. Eichhorn).

http://dx.doi.org/10.1016/j.bbcan.2017.09.002
Received 4 August 2017; Received in revised form 11 September 2017; Accepted 12 September 2017
Available online 18 September 2017
0304-419X/ © 2017 Elsevier B.V. All rights reserved.
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

Fig. 1. Copy number variation of deubiquitinating enzymes in cancer. Individual DUB expression in cancer subtypes extrapolated from cBioPortal (www.cBioPortal.org) derived from the
following datasets, [304–317], Adrenocortical Carcinoma (TCGA), Colorectal Adenocarcinoma (TCGA), Cervical Squamous Cell Carcinoma (TCGA), Uveal Melanoma (TCGA), Hepa-
tocellular carcinoma (TCGA), B-cell Lymphoma (TCGA), Ovarian adenocarcinoma (TCGA), Pancreatic Adenocarcinoma (TCGA), Skin Cutaneous Melanoma (TCGA), Testicular Germ Cell
Cancer (TCGA), Uterine Carcinosarcoma (TCGA). Expression was annotated in tumours if DUBs expression was shown to be altered in more than 5 individual cases.

direct transfer of ubiquitin from the E2 to the substrate. However, it has ubiquitin chains. Although the mechanism of chain formation is un-
been proposed that RING ligases may also function by luring ubiquitin known the majority of experimental evidence indicates that chains are
away from the E2 through allosteric mechanisms independent of its built sequentially on the substrate (Reviewed in Ye et al. [6]). However,
catalytic site [3,4]. The RBR family of ligases share many of the bio- a second mechanism of chain formation has been explored whereby
chemical features present in RING and HECT function including re- ubiquitin chains are sewn together on the E2's and E3's and transferred
cruiting ubiquitin conjugated E2 to RING1 domains. The bound E2 then as a single block to its cognate substrate.
transfers the ubiquitin to the second C-terminal RING domain whereby Polyubiquitin chain topology is determined by the varying ubiqui-
the ubiquitin bound at the intrinsic catalytic cysteine is transferred to tin–ubiquitin linkages which may occur. Ubiquitin chains can occur
the substrate. through specific K6, K11, K27, K29, K33, K48, or K63 linkages. Unlike
The transfer of ubiquitin to the substrate primarily occurs through HECT domain ligases where the transfer of ubiquitin to the substrate
the formation of a isopeptide bond between the carboxylic acid group of occurs directly from the HECTs own catalytic cysteine, the E2 ligase is
glycine 76 (Gly76) of ubiquitin to the epsilon amino group in the target accountable for the transfer of ubiquitin to the substrate when RING
lysine. Substrates may be modified at a single lysine residues (mono- ligases are involved. Therefore, it stands to reason that the type of
ubiquitnation) or at multiple lysine residues simultaneously (multi- ubiquitination that occurs is determined largely by the characteristics
monoubiquitination). Similarly, Gly76 of ubiquitin may attach to lysine of the E2. Many RING ligases can bind a number of E2s producing
residues on other ubiquitin molecules permitting the formation of poly- several types of ubiquitination products. This may explain the recent

457
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

observation that chain formation may either be homotypic (the links in Therefore, a better understanding of the dynamic interactions of DUBs
the chain all occur at a single lysine residue, for example K48) or het- and the pathways they regulate is essential. This review focuses on the
erotypic (the links in the chain may switch for example from K11 to tumour suppressor and oncogenic specific functions of E3 ligases and
K27). Furthermore, some ubiquitin chains have been described to DUBS in the major pathways involved in cancer progression including
contain SUMO moieties while others have exhibited a branched for- TGFβ, YAP, MAPK, PI3K, and WNT pathways. In reference to the role of
mation. All of this indicates that chain topology is eventually likely to ubiquitin modifying enzymes in the NFKB pathway we refer to the
be dependent on the three-dimensional interface between ligase and excellent review by Harhaj et al. [17].
substrate. The function of these distorted structures remains to be de-
termined but their discovery opens up a new window into the basic 2. The role of ubiquitin modifying enzymes in the TGFβ pathway
biology of ubiquitin function. Lastly, linear ubiquitin chains, also
known as M1 linked chains, can be generated through the formation of The Transforming Growth Factor β (TGFβ) pathway is crucial for
a covalent bond between the carboxy terminal glycine of ubiquitin and regulating embryonic development as well as maintaining tissue
the amino terminal methionine of the substrate bound ubiquitin [7]. homeostasis in adult tissues [18–20]. The majority of human cell types
It is well described that monoubiquitination and polyubiquitination are responsive to TGFβ, through which it regulates cell proliferation,
resulting from the various assortment of linkages described above differentiation, migration, adhesion, apoptosis and various aspects of
convey distinct structural and functional information. For the most part the microenvironment [18–20].
polyubiquitination is still epitomized by two ubiquitin chain topologies, TGFβ is a member of a large family of cytokines divided into two
K48 and K63. K48-linked chains serve to act as the prototypical de- distinct branches: TGFβ, activin, lefty, nodal and myostatin form one
gradation signal targeting the protein for proteasome mediated de- arm, whereas the other comprises of bone morphogenetic proteins
gradation while K63-linked chains perform a number of non-proteolytic (BMPs), anti-muellerian hormone (AMH) and other growth and differ-
functions including cellular signalling, DNA damage repair, in- entiation factors (GDFs) [19]. These classifications are simply based on
tracellular trafficking and ribosomal biogenesis. Due to the relative sequence homology and the distinct pathways that each activates [21].
infrequency of the other chain topologies they have been termed aty- Activation of the TGFβ pathway requires the ligand induced formation
pical linkages with their cellular functions remaining largely elusive of a tetrameric complex comprised of a pair of TGFβ receptor I (TβRI)
[8]. subunits as well as a pair of TGFβ receptor II (TβRII) subunits [22].
Similar to the way that protein phosphatases oppose the action of TβRII is a constitutively active serine/threonine kinase and upon ligand
protein kinases, ubiquitination is a process that is also reversible, binding it is able to phosphorylate TβRI at a distinct location preceding
having a family of enzymes that counteract the role of the ligase. the kinase domain called the GS domain [23]. This results in the acti-
Ubiquitin moieties can be removed from polypeptides by DUBs. The vation of the TβRI serine/threonine kinase allowing for effector mole-
human genome encodes approximately 80 DUBs which are grouped cules, Receptor–SMADs (R-SMADs), to be phosphorylated [18–20]. The
into six distinct families: ubiquitin-specific protease (USP, 57 mem- R-SMADs belonging to the TGFβ arm of the pathway include SMAD2
bers), ubiquitin C-terminal hydrolase (UCH, 4 members), otubain pro- and SMAD3, whereas SMAD1, SMAD5 and SMAD8 are attributed to the
tease, (OTU, 14 members), motif interacting with Ub-containing novel BMP pathway [18,19].
DUB family (MINDY, potential 4 members, only 1 is discuss in this SMAD proteins are made up of two globular Mad-homology (MH)
review), and machado-joseph disease protease (MJD, 4 members). USPs domains, MH1 and MH2, separated by a highly regulated linker region
make up by far the largest family of these thiol proteases. The sixth DUB [24]. The MH1 domain contains the conserved DNA binding motif
family, is distinct from the other 5 DUB families in that they possess a capable of recognizing the 51-AGAC-31 DNA sequence, more commonly
JAMM (JAB1/MPN/Mov34 metalloenzyme, 11 members) domain to known as the SMAD-Binding-Element (SBE). MH2 is highly conserved
carry out their catalytic function [9,10]. The deubiquitination ability and mediates complex binding with other SMADs or SMAD nuclear
and substrate specificity of a particular DUB is determined by chain complexes. The variable linker region is enriched in proline residues
topology, sub cellular localization and protein complex formation. It is and contains multiple phosphorylation residues which can be phos-
feasible that any disequilibrium in the ubiquitination-deubiquitination phorylated by various kinases in response to stimuli such as MAPK, or
balance can act as a major contributing factor to tumour formation. To CDK8/9 [25].
further understand the potential role of DUBS in tumourgenesis we R-SMADs are phosphorylated at a precise-SSXS c-terminal motif
performed expression analysis of all the known DUBs across 20 tissue creating an interaction interface permitting the association with the co-
specific datasets using cBioPortal (Fig. 1). Our analysis reveals that SMAD, SMAD4. Upon entry to the nucleus the R-SMAD/co-SMAD
across many tumour types, there appears to be a general trend towards complex binds to the SMAD binding element (SBE) sequence on DNA
DUB amplification. Given that gene copy number alterations are a permitting appropriate transcriptional responses [18,20,21].The R-
common phenomenon in cancer our observations could merely be a SMAD/co-SMAD complex has only a weak affinity for the SBE response
result of this. It is, however, conceivable that the overexpression of K48 elements, thus full transcriptional activation requires the association of
specific DUBs, by virtue of stabilizing oncogenes, could potentially be additional transcription factors [18–21]. This deceptive weakness in
critical determinants in tumorigenesis. Undoubtedly, a much more in- this family of transcription factors appears to be overcome by the
depth analysis will be required before this can be claimed with any overall flexible nature of the linker region whereby it is thought that the
certainty. The exception to this is observed in brain and prostate cancer distinct phosphorylation patterns in the R-SMAD linker region are likely
where deletion of a number of DUBs facilitate tumorigenesis suggesting to result in varying conformational differences which will allow the
that in these cancer types these DUBs operate as potential tumour various permutations of available transcription factors and the sub-
suppressors. Nevertheless, in the majority of cases DUB expression and sequent recruitment of co-repressors or co-activators to dictate whether
function (tumour suppressor or oncogene) appears to be tissue and a gene will be repressed or activated, respectively, by TGFβ stimulation
context specific. For example, USP9X has been shown to stabilize the [18–20]. Therefore, it is the versatility of the R-SMAD/co-SMAD com-
oncogenes β-catenin, MCL-1, estrogen receptor, Ets-1, among others, plex in binding a variety of other transcription factors which accounts
promoting cancer progression [11–14]. In contrast, USP9X expression is for the pleiotropy response to TGFβ.
lost in renal cell carcinoma and pancreatic ductal adenocarcinoma with Given the fundamental cellular processes that TGFβ regulates one
downregulation of USP9X corresponding with increased YAP and RAS can speculate that should this signalling pathway be deregulated,
signalling, respectively [15,16]. This apparent contradiction in function aberrations in cellular function would therefore occur promoting tu-
has restricted the development of novel inhibitors targeting DUBS and morigenesis. To investigate the validity of this, one must first under-
limited the progression of DUB inhibitors through clinical development. stand how TGFβ signalling is regulated. As this review will reveal, the

458
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

Fig. 2. Schematic overview of regulation of TGFβ pathway by Ubiquitin ligases and deubiquitinating enzymes. (A) Factors that turn off the TGFβ pathway: At the receptor level AIP4/
Itch, in a non-catalytic manner, brings SMAD7 and TGFβRI into close proximity preventing the association of SMAD2 with the latter and thus prevents signalling. SMAD7 itself brings
HECT E3 ligases SMURF1, SMURF2, Tuil1/WWP1 and NEDD4-2 to the receptor complex resulting in complex ubiquitination and degradation. The DUB, USP26, facilitates this event by
increasing the stability of SMAD7 by removing degradative ubiquitin chains. TGFβ pathway signal mediators are also regulated by degradative ubiquitin: SMURF2 targets SMAD2 for
degradation whereas SMAD3 is targeted by U-Box E3 ubiquitin ligase, CHIP; HECT E3 ligase, NEDD4L; and SCF E3 ligase ROC1-SCFFbw1a. Co-SMAD, SMAD4, is regulated by a number of
HECT E3 ligases, namely: SMURF1, SMURF2, Tuil1/WWP1, and NEDD4-2. These ligases associate with and ubiquitinate SMAD4 indirectly by forming a complex with SMAD7, SMAD6 or
activated SMAD2, resulting in the degradation of the former. In many cancers SMAD4 harbors point mutations. In this context these protein variants are targeted for degradation by SCF
E3 ligases, SCFTrCP1β and SCFSkp2. In addition to promoting degradation, ubiquitination is also able to disrupt protein complexes: SMURF2 multi-monoubiquitinates SMAD3 in either the
cytoplasm or the nucleus resulting in the dissociation of the R-SMAD/SMAD4 complex and hence disruption of TGFβ signalling. A similar reaction occurs in the nucleus as E3 ligase
Ectodermin/Tif1γ monoubiquitinates SMAD4 also resulting in the dissociation of the R-SMAD/SMAD4 complex. Once the R-SMAD/SMAD4 complex enters the nucleus to initiate
transcription, here SMAD2 is targeted for degradation as repressive DNA binding proteins SnoN and TGIF direct NEDD4-2 and Tiul/WWP1, respectively, to this R-SMAD, resulting in the
termination of signalling. It has also been shown that the action of an undetermined ligase is able to mono-ubiquitinate the R-SMADs in the R-SMAD/SMAD4 complex thus precluding
complex binding to the DNA. (B) Factors the turn the on the TGFβ pathway: At the receptor level, USP4 is able to interact directly with TGFβRI resulting in its relative persistence at the
plasma membrane level therefore promoting TGFβ signalling. DUBs USP11, USP15 and UCH37 all augment TGFβ signalling by associating with SMAD7, which effects a plasma
membrane localization and therefore stabilizes the receptor complex by way of degradative-ubiquitin removal. SMAD7, itself an antagonist to TGFβ signalling, is degraded by ubi-
quitination by three difference ligases: Arkadia, AIP4/Itch and RNF12. Further downstream, AIP4/Itch has also been observed to ubiquitinate SMAD2, resulting in an augmentation of
TGFβ signalling. The mechanism for this is unclear but this ubiquitination ostensibly promotes SMAD2 and TGFβRI proximity as an AIP4/Itch, SMAD2 and TGFβRI trimeric complex have
been observed in TGFβ stimulated cells (not depicted). Stability of SMAD2/3 is enhanced by the action of OTUB1, which prevents E2 ligase transfer of ubiquitin to the R-SMADs. USP9X
promotes the formation of the R-SMAD/SMAD4 complex by removing the ubiquitin moiety that is attached to SMAD4 by Ectodermin/Tif1γ, therefore relieving steric inhibition. Once in
the nucleus the R-SMAD/SMAD4 complex facilitates the transcription of TGFβ target genes by inducing the destruction of transcriptional repressor SnoN: Arkadia, SMURF2 and CDH1-
APC bind to SMAD2/3 in the nucleus bringing the ligases into close proximity to SnoN allowing for ubiquitin mediated degradation. Finally, as mono-ubiquitination of R-SMADs
precludes the R-SMAD/SMAD4 complex from associating with the DNA, USP15 therefore promotes TGFβ dependent transcription by removing this ubiquitin moiety.

dynamic ubiquitination/deubiquitinating process plays a crucial role in found to be a ubiquitin ligase for SMAD1 resulting in the degradation of
regulating overall TGFβ signalling at all nodes of the pathway (Fig. 2). SMAD1 and loss of BMP signalling [27]. Subsequent analysis found,
It will also provide pertinent examples of how aberrations in this pro- however, that it did not bind to SMAD2, indicating that another E3
cess can lead to cancer formation. ligase was responsible for the negative feedback mechanism originally
identified by Lo and Massague [28]. The SMURF1 homologue, SMURF2
2.1. E3 ligases in the TGFβ pathway was subsequently shown to bind SMAD2 in a TGFβ dependent manner,
targeting SMAD2 for proteasomal degradation [29,30]. However, both
The earliest observation that TGFβ signalling is regulated by ubi- Bonni et al. and Zhang et al. contest the notion that SMURF2 can de-
quitination was in 1999 by Lo and Massague where they observed that grade SMAD2 with Zhang et al. conceding that SMURF2 can degrade
prolonged TGFβ exposure reduced levels of total SMAD2 in a protea- SMAD2 but only when ectopic expression of SMURF2 is high [31,32]. It
some dependent manner. Their discovery has led to almost 20 years of is important to note that it remains inconclusive if endogenous SMAD2
research in ubiquitin dependent regulation of the TGFβ pathway [26]. can be degraded by SMURF2 [32,34]. Both SMURF1 and SMURF2 are
With regard to ubiquitin mediated regulation the TGFβ superfamily as a C2-WW-HECT-domain ligases capable of binding SMADs through an
whole, the first major discovery was the identification of the HECT E3 intermolecular interaction between the SMURFs WW domain and the
ligase SMAD-specific E3 ubiquitin-protein ligase 1 (SMURF1). This was PPXY sequence (PY) motif in SMADs [35]. SMURF1 has also been

459
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

shown to ubiquitinate and degrade SMAD5, while SMURF2 also ubi- of the TβR complex hence removing the tumour suppressor effect of
quitinates SMAD1 under steady-state conditions [32,36]. TGFβ signalling [44].
NEDD4-like or NEDD4-L is another well-studied E3 ligase, which Yet another HECT E3 ligase, NEDD4-2 (neural precursor cell ex-
belongs to the same E3 ubiquitin ligase family as the SMURF proteins. pressed developmentally down-regulated 4-2), has been found to ubi-
NEDD4-L specifically recognizes a threonine phosphorylation site di- quitinate and degrade SMAD2 [46]. This ubiquitination and degrada-
rectly prior to the PY motif in the linker region of R-SMADs allowing tion occurs in the presence of activated TGFβ signalling and since
protein-protein recognition and marking SMAD2/3 for proteasomal NEDD4-2 can bind nuclear TGFβ repressor SnoN (see later regarding
degradation. Ostensibly, therefore certain phosphorylation residues in SnoN) via its association with SMAD2 this suggests that this process
the linker region enhance SMAD transcriptional action before being again takes place in the nucleus [46]. NEDD4-2 therefore may con-
marked for degradation by NEDD4L. tribute along with Tiul1 to the negative feedback instigated due to
Controversially, recent results by Tang et al. clearly indicate that prolonged TGFβ exposure.
SMURF2 does not regulate protein stability of these aforementioned It has been observed that SMAD3 is also regulated by ubiquitina-
SMAD proteins. Intriguingly, they demonstrate that in engineered tion. This ubiquitination is mediated by the RING E3 ligase ROC1-SCF-
Smurf2 (−/−) mouse embryonic fibroblasts turnover rates of SMAD2/ Fbw1a where this complex binds the MH2 domain of SMAD3 [47]. SCF
3 were equivalent compared to wild type fibroblasts. However, loss of complexes are comprised of three proteins, namely, SKP1, Cullins and
SMURF2 did appear to alter monoubiquitination at multiple lysine re- F-box proteins, where F-box proteins are important for substrate spe-
sidues in the MH2 domain of SMAD3. Monoubiquitination of the K333, cificity [48]. In the aforementioned complex, a RING finger protein
K378, and K409 in the MH2 domain of SMAD3 blocks formation of both ROC1 associates with the SCF and is crucial for the ubiquitination
homotrimeric SMAD3 and heterotrimeric SMAD3-SMAD4 complexes function of the entire complex [47]. The degradation of SMAD3 by
invariably limiting these complexes in binding to SMAD motifs on the ROC1-SCF-Fbw1a is TGFβ dependent. Interestingly, SMAD3 is exported
DNA [37]. The interesting finding that SMURF2 targets SMAD3 for out of the nucleus by ROC1-SCF-Fbw1a where it is presumed that in the
monoubiquitination is in line with recent results that show that NEDD4 cytoplasm degradation occurs [47]. This is in contrast to what is ob-
family members (of which SMURF1 and SMURF2 are included) pre- served with SMAD2, as mentioned earlier. Of note, it has been subse-
dominantly assemble K63 linkages [38]. The fact that SMURF proteins quently found that U box E3 ligase, CHIP, is able to target SMAD3 ir-
may only form K63 chains and not K48 chains as was once believed respective of phosphorylation status [49].
leads to a host of questions regarding the role of SMURF2 targeting In addition to targeting the R-SMADs, ubiquitin based regulatory
proteins for degradation. It may be that SMURF proteins may only mechanisms also exist for co-SMAD, SMAD4. Here again we see that a
prime their targets for entry into early endosomes whereby other ligases negative feedback mechanisms are built in to the regulation of this
targeted these proteins for degradation. On the other hand, SMURF protein [50]. When TGFβ ligand is added, the F-box protein β-TrCP1
proteins might function in a dual role: priming targets through mono- putatively recognizes a phosphorylated “DLSGLTLQS” motif on SMAD4
ubiquitination and later catalysing polyubiquitinated chains depending resulting in the degradation of the latter [50]. Post translational mod-
on other regulatory proteins bound in the complexes. ification of this motif remains unproven but it has been postulated that
Tiul1/WWP1 is another member of the HECT E3 ligase family and Jun activation domain binding protein 1 (JAB1) may promote phos-
has also been found to promote degradation of SMAD2 [39]. Upon phorylation of SMAD4 at this motif [50]. Importantly, JAB1 has been
stimulation with TGFβ, Tiul1/WWP1 interacts with Transforming observed to interact with and induce degradation of SMAD4 [51].
growth factor beta-inducing factor 1 (TGIF) and facilitates the forma- SMAD4 is prominently inactivated at the genomic level in pancreatic
tion of a repressor complex eventually leading to the ubiquitination and cancer and is considered a key driver for tumorigenesis in this context
degradation of SMAD2 [40]. Lo and Massague have previously ob- [52,53]. Furthermore, SMAD4 point mutations isolated from pancreatic
served that SMAD2 is degraded specifically in the nucleus. This ob- tumours appear to be correlated with errant ubiquitination patterns,
servation coupled with the fact that Tiul1/WWP1 mediated degradation and overall increased protein turnover [54]. This is most likely due to
of SMAD2 is induced by prolonged TGFβ ligand (16 h) suggests that this misfolding of the mutant protein and subsequent ‘quality control’ ubi-
ligase may be responsible for the TGFβ induced negative feedback loop quitination [54]. Furthermore F-box protein SKP2 which has been de-
first observed by Lo and Massague [28]. monstrated to preferentially bind and degrade cancer derived SMAD4
Importantly, in breast cancer the chromosomal band 8q21 is often mutants [54]. It interesting to note here that unlike SMAD4, SMAD2
amplified [41]. Tiul1/WWP1 gene is present within this band and it and SMAD3 are rarely mutated in cancer suggesting that this type of
was found that copy number gain was present in approximately 51% of ubiquitin mediated degradation of SMAD2/3 is unlikely to operate in
breast cancer cell lines and 41% of primary breast tumours [41]. As cancer [19].
already described Tiul1/WWP1 is a negative regulator of TGFβ sig- Degradative poly-ubiquitination of SMAD4 can also be mediated by
nalling and further biochemical analysis confirmed that siRNA tar- various HECT E3 ligases, including SMURF1/2, WWP1 and NEDD4-2
geting of Tiul1/WWP1 in breast cancer lines enhanced TGFβ mediated [54]. It has been proposed that these ligases do not directly interact
responses such as cytostasis and apoptosis [41]. This implies that ele- with SMAD4 but rather regulate SMAD4 physiological levels through
vated Tiul1/WWP1 provides an early proliferative advantage in breast complex formation with either inhibitory SMADs (I-SMADs) or SMAD2
cancer and in particular, in estrogen receptor positive breast cancers [54]. As this interaction appears to occur in the cytoplasm this may
[41,42]. A similar phenomenon is observed in prostate cancer with 44% suggest a mechanism of quashing latent SMAD4 which can no longer be
of prostate cancer xenografts and cell lines and 31% of prostate clinical recycled following re-entry into the cytoplasm from the nucleus, thus
samples possessing a gain in Tiul1/WWP1 copy number [43]. Similar to maintaining tolerable SMAD4 levels quantities for R-SMAD/Co- SMAD
breast cancer, siRNA targeting of Tiul1/WWP1 in a prostate cancer cell binding. Conversely, Ectodermin/Tif1γ (ECTO) was identified to be a
line enhanced TGFβ induced cytostasis, again implying that over- mono-ubiquitin ligase for SMAD4 specifically ubiquitinating at the
expression of Tiul1/WWP1 has oncogenic capacity [43]. In addition, K519 site thus preventing the formation of SMAD4/R-SMAD4 hetero-
hyperactivity of Tiul1/WWP1 has also been noted in prostate cancer trimer complex and inhibiting chromatin binding [57]. With ubiquiti-
[44].To limit the intrinsic autocatalytic activity inherent to HECT E3 nation occurring in the nucleus the monoubiquitinated form of SMAD4
ligases intramolecular interactions between the C2 and HECT domains is exported to the cytoplasm. Interestingly, it has been hypothesized
maintain the ligase in closed inactive confirmation [41,44,45] Inter- that the acetylation of histones in close proximity to the chromatin
estingly, prostate cancer derived point mutant WWP1- E798V was bound SMAD complexes increases the affinity for ECTO thus expelling
found to be hyperactive due to the disruption of the aforementioned the SMAD complex from the promoter regions on the DNA resulting in
auto-inhibition mechanism. This disruption lead to hyper-degradation the loss of transcription of downstream TGF-β target genes.

460
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

In addition to E3 ligases acting downstream in the TGF-β pathway, negative regulators for proteasomal degradation including, SMAD7, c-
they also mediate TGF-β kinetics at the receptor level. As part of a Ski, and SnoN [73,74]. As previously mentioned HECT E3 ligases bind
transcriptionally regulated negative feedback loop BMP and TGFβ sig- to PY domain of SMAD7, in contrast, ARKADIA binds to the MH2 do-
nalling induces the expression of the inhibitor adaptor SMADs (I-SMAD) main of SMAD7 and induces the polyubiquitination and degradation of
SMAD6 and SMAD7, respectively. I-SMADs are distinct from the R- SMAD7 leading to enhanced TGFβ signalling [74]. This uncovers per-
SMADs in that they possess weak homology with respect to the SMAD1 haps an interesting insight to how whereabouts of binding to SMAD7
MH1 domain [58–60]. The function of these SMADs is principally to influences whether a particular ligase will degrade SMAD7 or simply
alleviate TGFβ output [58–60]. SMAD6 has been shown to associate uses it as an adaptor protein.
with type 1 receptors TβRI and BMP receptor IB (BMPRIB), mitigating C-Ski and SnoN are related transcriptional co-repressors which as-
the levels of p-SMAD2 and p-SMAD1, respectively [59]. It has also been sociate with the R-SMAD-SMAD4 complexes and recruit histone dea-
reported that SMAD6 inhibits BMP signalling by competing with cetylases (HDACs) to prevent transcription of TGFβ target genes [31].
SMAD1 for physical association with SMAD4 [61]. SMAD7 has two SnoN levels are partially regulated through ligand induced phospho-
distinct mechanisms by which it inhibits TGFβ signalling. Firstly, SMAD2 whereby phospho-SMAD2 translocates to the nucleus forming a
SMAD7 binds preferentially to activated TβRI hence competing with R- complex with ARKADIA targeting SMAD2 bound SnoN for ubiquitin
SMADs and subsequently mitigating pathway activation [58]. However, mediated degradation. Interestingly, ARKADIA binds and ubiquitinates
a potentially more important role for SMAD7 appears that it is the key SnoN in the absence of TGFβ signalling [76]. Nevertheless, it is only
node for ubiquitin mediated regulation of the TGF-β pathway. Me- upon TGFβ pathway activation that efficient ARKADIA induced SnoN
chanistically, SMAD7 serves as scaffold to recruit SMURF2 and the E2 degradation occurs due the formation of a phospho-SMAD2 -SnoN-
ligase UBCH7 to the TGF-β receptor complex to facilitate receptor ARKADIA complex [76]. This insinuates that phospho-SMAD2 is re-
polyubiquitination and complex degradation [62]. quired for proteasome targeting [76]. Interestingly, under conditions
The interaction of SMAD7 to SMURF2 also has a secondary function. where ARKADIA expression is repressed phospho-SMAD2/3 accumu-
As previously mentioned, SMURF2 possesses autocatalytic activity lates in the nucleus but appears to be non-functional. Furthermore,
when the protein is unfolded and therefore to maintain its stability and reconstitution of ARKADIA in null embryonic stem (ES) cells results in a
constrain unwanted activity towards its substrates the C2 and HECT restoration of SMAD2/3 transcriptional activity. However, counter-
domains remain in a tightly closed confirmation. The binding of SMAD7 intuitively, under these conditions phospho-SMADs gets degraded in
to the HECT domain of SMURF2 abrogates these inhibitory in- the process [77]. This coupling of activation with degradation has been
tramolecular interactions between these domains facilitating SMURF2 suggested to provide a mechanism to regulate appropriate transcrip-
ubiquitin ligase activity [63]. Besides acting as a scaffold for SMURF2 it tional responses to ensure that active transcription is transient and that
performs a similar function for the E3 ligases NEDD4-2, SMURF1, and transcription factors do not perpetually remain on promoter sequences.
its homologue WW Domain Containing E3 Ubiquitin Protein Ligase 1 Thus, once transcription has been initiated utilized SMAD complexes
(WWP1) also known as Tiul1 (TGIF interacting ubiquitin ligase 1), will be targeted for degradation or recycling, allowing new SMAD
which also target the type I TGF-β receptors for ubiquitin mediated complexes to bind to the DNA. This termination signal is then one
degradation [39,64–66]. The existence of this apparent redundancy in mechanism to ensure that the level of extracellular TGFβ ligand pro-
function between the HECT E3 ligase family is unclear and may ulti- duces the desired physiological responses. Whether the accumulation of
mately be context dependent. Furthermore, CD109 also binds to the nuclear SMADs in ARKADIA null ES cells is the result of increased SnoN
SMURF2/SMAD7 complex and regulates receptor ubiquitination and expression and the inhibition of binding of SMAD complexes to DNA
degradation in a ligand dependent manner [67]. remains inconclusive but is likely to be part of the process.
It has been observed in renal cell carcinoma that TGFβ signalling is Nagano et al. report that ARKADIA can bind and degrade SnoN, as
depressed when compared with normal renal tissues, with a decrease in well as c-Ski, a SnoN related repressor, independently of TGFβ signal-
signalling being correlated with a post-translational decrease of TβRII ling, contrasting previous observations [73,78]. Incidentally SMURF2
protein levels [68]. In fact, in renal cell carcinoma tissues the levels of has also been demonstrated to target SnoN via SMAD2 [79]. SMAD2 is
SMURF2 protein are inversely correlated with TGFβRII [68]. High le- essential for this process as the utilization of SMAD2 deleted for its PY
vels of SMURF2 also correlate with poor prognosis in Esophageal motif limited the ability of SMURF2 to bind SnoN [31].
Squamous Cell Carcinoma [69]. It was found that in this cancer, Interestingly, SMAD3 also functions to degrade SnoN but in this
SMURF2 is overexpressed compared with normal tissues and this high scenario degradation occurs via the RING E3 ligase, Anaphase
expression was correlated with depth of invasion, and lymph node Promoting Complex (APC)-CDH1 complex. CDH1 acts as an adaptor for
metastasis [69]. Unlike renal cell carcinoma, the main substrate of specific substrate recognition [80]. It is reported that this action is also
SMURF2 in this context appeared to be pSMAD2. TGFβ dependent as APC only interacts weakly with SnoN in the absence
Atrophin 1-interacting protein 4/ITCH(AIP4/Itch) subsequently to of ligand. This can be explained by the fact that destruction box motif
be referred to as ITCH is another HECT E3 ligase that acts to inhibit (D- Box) within SnoN, which the CDH1 subunit recognizes, is not per-
TGFβ signalling [70]. In contrast to other members of the HECT family, fect thereby limiting CDH1-SnoN affinity [80]. SMAD3 is therefore
in this context, ITCH operates independently of its ligase activity [70]. required for efficient association of the APC complex with SnoN [80].
ITCH appears to strengthen the association between SMAD7 and acti- Recalling the observation made by Bonni et al. that phospho-SMAD2 is
vated TGFβRI (without affecting turnover of TGFβRI) and in this way required for SMURF2 recruitment to degrade SnoN, employing a
blocks R-SMAD access to the receptor kinase domain [70]. In a con- SMAD3 mutant incapable of SMURF2 binding similarly led to reduced
trasting study, Bai et al. observed that ITCH enhances pSMAD2 levels degradation of SnoN [79,80]. This suggests that both SMURF2 and APC
by ubiquitination of SMAD2. They observe the formation of a trimeric mediated degradation of SnoN operates concurrently for maximal effect
complex between ITCH, SMAD2 and TGFβRI in stimulated cells and [80]. The contextual importance of the TGFβ negative regulator
therefore postulate that this ubiquitination promotes SMAD2 proximity SMURF2 repressing SnoN, another negative regulator remains to be
with TGFβRI thus facilitating overall signalling [71]. In addition, ITCH determined.
has also been shown to act as a positive regulator of the TGFβ pathway ARKADIA may also be involved in endocytosis of TβR as it has been
by ubiquitinating and inducing degradation of SMAD7 [72]. It is un- shown to bind and ubiquitinate the μ2 subunit of AP-2, which is in-
clear why ITCH facilitates such opposing effects with respect to TGFβRI volved in the formation of clathrin coated pits [81]. ARKADIA's overall
activity but it is likely to be context dependent. importance in terms of cancer remains inconclusive. Although the loss
The ubiquitin-proteasome system can also positively regulate the of ARKADIA is rare in human cancers a number of missense mutations
TGFβ cascade. ARKADIA, a RING-finger containing E3, targets multiple have been localized in colorectal cancer patients correlating with SnoN

461
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

stabilization thus potentially confirming the role of ARKADIA as an of course leaves us with the question of what is the E3 ligase that
tumour suppressor [82] [83]. Furthermore, mice heterozygous for monoubiquitinates the lysine residues within the MH1 domain.
ARKADIA possess tumours with enhanced nuclear SnoN and decreased The fact that TGFβ enhances USP15 proteins levels but at the same
TGFβ signalling leading to colorectal cancer development [82]. Finally, time dissociates USP15 from the SMAD7/SMURF2 complex is in intri-
it has been found that in the esophageal cancer cell line (SEG1) which guing enigma. Recently, it has been demonstrated that USP15 binds to
exhibits low levels of ARKADIA expression, degradation of SnoN is also and stabilizes the transcription factor p53 [89]. Under physiological
subsequently impaired [76]. conditions p53 can form complex with SMAD2/3 to co-activate tran-
In addition to activation of R-SMADs through the canonical TGFβ scription of an assortment of genes involved in tumour suppression. It
signalling pathway, TGFβ also the recruits tumour-necrosis factor re- may therefore be that under certain conditions USP15 acts as a general
ceptor (TNFR)-associated factor TRAF4 to the TGFβ receptor complex regulator of the TGFβ tumour suppressor function by deubiquitinating
restraining the E3 ligase activity of SMURF2 towards the receptor both R-SMADs and its co-transcriptional regulator p53 enhancing TGFβ
complex thereby limiting ubiquitination and degradation and thus mediated transcription of genes. However, in scenarios where p53 is
maintaining TGFβ activity. Furthermore, TRAF4 is also able to recruit mutated or lost and the levels of USP15 are abnormally high, this may
the deubiquitinating enzyme USP15 to aid in this process (see below). switch the binding of SMAD transcription factors from the promoters of
RNF12 has also been identified as a ligase for SMAD7 degradation [84]. tumour suppressor genes to those governing more pro- proliferative
functions. This appears to be the case in certain cancers as USP15 gene
2.2. DUBs in the TGFβ pathway was demonstrated to be amplified in glioblastoma, breast and ovarian
cancers Using a number of independent samples sets, Eichhorn et al.
Considering the importance of the TGFβ pathway it is not surprising was able to confirm that USP15 expression is highly correlated with
that TGFβ signalling is heavily regulated by the ubiquitination process, TGFβRI and pSMAD2 in these cancers. Furthermore, glioblastoma pa-
which is involved in crucial negative feedback loops, quality control tients with relatively high copy numbers of USP15 have a significantly
mechanisms and constitutive degradation to maintain homeostasis. poorer prognosis compared to those with lower copy numbers [91].
Ubiquitination is a reversible process and this adds another level of As previously mentioned a number of DUBs including USP4, 11, and
regulation to the TGFβ pathway. This reversal process mediated by 19 have been identified as potent regulators of the signalling cascade.
deubiquitinating enzymes (DUBs) provides further opportunities for All of these DUBs bound to the TβRI directly resulting in decreased
aberrations to arise and facilitate cancer progression. levels of TβRI ubiquitination and stabilization. The existence of DUBs
TβR stability and turnover functions as a principle juncture for the with apparent overlapping functions can only attest to the level of
downregulation of the pathway. Counteracting the ubiquitination and complexity required for internalization and compartmentalization of
degradation of the TβR complex six DUBs (USP4, USP11, USP15, receptors at the cell surface. Interestingly, the fate of TGFβR is de-
USP19, and UCH37) have been identified that directly affect TβR pendent upon a continuous interplay between ligases and DUBS to
deubiquitination and stability [62,85–87]. Making use of genome wide regulate ubiquitination and internalization. This is not only determined
DUB libraries three independent groups identified USP15 as a critical by transitory ubiquitination state of the receptor but of all components
component of the TGF-β pathway. Interestingly, akin to many of the E3 of the receptor complex including the ligases and DUBs themselves.
ligases USP15 appears to target a number of different nodes in the TGFβ Preliminary results by Ten Dijke and colleagues add weight to this
pathway. At the receptor level USP15 forms a complex with SMAD7 and theory whereby they identified that the ability of USP15 to deubiqui-
SMURF2 with USP15 opposing the effects of the SMURF2 ligase on TβR tinate TβRI requires USP4, as USP15 was unable to perform this func-
stabilization [87,88]. In this context, the scaffold protein SMAD7 in- tion in USP4 deficient cells [85]. Furthermore, they demonstrate that
teracts with two enzymes harbouring contrasting activities resulting in AKT phosphorylation of USP4 enhances the binding of USP4 to USP15
a constant balancing act regulating TGF-β output. Recently, it has been and that overexpression of USP15 increases USP4 stability. Under these
shown that TGFβ upregulates the translation of USP15 suggesting a conditions AKT hijacks USP4 to stabilize the TβRI and reinforces the
positive feedback loop, however, the extent of the TGFβ activity also pro-tumorigenic responses of the TGFβ pathway. Notably, USP4 is
regulates the access of USP15 to the SMAD7-SMURF2 complex [87,89]. overexpressed in breast cancer and may function as an oncogene to
As a result, when the TGFβ signal is low SMAD7 engages both SMURF2 maintain the TGFβ breast cancer progression [92]. Regarding the cross
and USP15 to maintain TβR stability thus retaining low levels of TGFβ talk observed between enzymes regulating ubiquitination and inter-
output. However, when excessive levels of TGFβ is present USP15 is nalization of receptors it would be of great surprise if AKT did not
dissociated from the SMAD7-SMURF2 complex leading to enhanced regulate other components of this complex.
ubiquitination of the TβR and degradation of the complex. This gen- Not unlike USP15, two other DUBS USP11 and UCH37 are able to
erates an elegant rheostat whereby TGFβ regulates its own activity bind SMAD7 and target the TGFβ receptor for deubiquitination [62,93].
preventing hyperactivation of the signal cascade. In the case of USP11, SMAD7 does not appear to act exclusively as a
USP15 also appears to regulate other nodes within the canonical scaffold for as SMAD7 does not appear to mediate the interaction be-
TGF-β pathway. As well as acting as a DUB for the TβR, USP15 has tween USP11 and TβRI, despite the fact that SMAD7 and USP11 being
recently been identified to deubiquitinate monoubiquitinated and able to form a complex. In addition, the interaction between USP11 and
polyubiquitinated isoforms of R-SMADs [90]. Post translational mod- SMAD7does not appear to be greatly affected by TGFβ ligand. However,
ifications within the DNA binding domains of R-SMADs prevents pro- besides acting as TβRI deubiquitinases both USP15 and USP11 are able
moter recognition and aberrant TGFβ signalling. As previously dis- to regulate their counterpart the E3 ligase SMURF2 albeit through ap-
cussed, Tang et al. demonstrated that SMURF2 knockout MEFs, rather parently divergent mechanisms [88]. USP15 appears to target key re-
than displaying the expectant stabilization of known substrates merely sidues within the HECT domain of SMURF2 thereby limiting the cata-
exhibited decreased mono-ubiquitinated isoforms of SMAD3 [37]. lytic activity of SMURF2 towards it's substrates, while USP11 appears to
These specifically occurred at K333, K378, and K409 within the MH2 increase overall SMURF2 ubiquitination. The effect of USP11 in this
domain. In contrast, USP15 deubiquitinates mono-ubiquitinated context is as of yet undetermined but it may suggest that USP11 may
SMAD3 at K81 and to a lesser degree at K33 and K53 suggesting that indirectly regulate SMURF2 stability.
USP15 deubiquitinates R-SMADs in the MH1 domain at sites in- All the DUBs mentioned thus far, bar USP4 and USP11, utilize
dependent of SMURF2 function [90]. Notably, all of these ubiquitina- SMAD7 as a scaffold protein to mediate their effects. Recently, we
tion sites disrupt the R-SMAD/SMAD4 heterodimer from binding to the identified that as part of a negative feedback loop TGF-β not only en-
chromatin. USP15 reverses this modification permitting SMAD tran- hances the expression of SMAD7 but also of USP26, whereby USP26
scription factor binding and full TGF-β transcriptional responses. This acts as a SMAD7 K48 ubiquitin chain specific DUB rescuing SMAD7

462
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

Fig. 3. Schematic overview of regulation of Hippo pathway by Ubiquitin ligases and deubiquitinating enzymes. Activation of the Hippo pathway results through multiple extracellular
mediums including cell-cell contact or mechanical tension leading to the downstream activation of two key core inhibitory kinase modules. The first of these comprises of MST1 and MST2
and the second module comprises of LATS1 and LATS2 (Drosophila Hippo and Warts, respectively). MST1/2 in combination with its binding partner Sav1 permits phosphorylation and
kinase activation of LATS1/2 and the LATs co-factors MOB1A and MOB1B. Downstream transcription is mediated by YAP and its paralogue TAZ. These two transcription factors shuttle
back and forth between the cytoplasm and the nucleus where they regulate transcription of a multitude of genes primarily through TEA domain family members (TEAD). LATS1/2
phosphorylation of the YAP/TAZ complex displaces YAP/TAZ to cytoplasm inhibiting YAP/TAZ mediated transcription. Phosphorylation of YAP/TAZ by LATS1/2 occurs in a unique
phosphodegron site inducing the recruitment of the E3 ligase β-TrCP resulting in polyubiquitnation and degradation of YAP/TAZ. Similar to LATS1/2, Angiomotin (AMOT) regulates
hippo pathway activation by directly binding to YAP/TAZ resulting in cytoplasmic retention of both proteins. Ubiquitination regulates numerous proteins in the hippo pathway. AMOT
itself undergoes polyubiquitination and degradation by the E3 ligases NEDD4 and ITCH inhibiting AMOT regulation of YAP/TAZ. USP9X has been shown revert this effect. NEDD4, ITCH,
its HECT family member WWP1, and SIAH2 also ubiquitinate and degrade LATS1/2, and NEDD4 has also been demonstrated to perform a similar function towards SAV1. The E3 ligase
PRAJA2 ubiquitinates and inhibits the stability of MOB1. All of which result in downregulation of hippo pathway activation and enhanced YAP/TAZ transcription. The deubiquitinating
enzyme DUB3 deubiquitinates and stabilizes multiple components of the hippo pathway including the negative regulator ITCH resulting in AMOT and LATS1/2 degradation. Remarkably,
DUB3 can also deubiquitinate AMOT and LATS enhancing hippo activity.

from degradation. This then permits SMAD7 to remain in stable con- By modulating the activity of co-SMAD, SMAD4, USP9X/FAM has
firmation with SMURF2, permitting SMURF2 recruitment to the TGF-β also been found to modulate the TGFβ pathway [57]. To counteract the
receptor complex potentiating complex degradation [94]. Other I- monoubiquitination of SMAD4 by Ectodermin at K519 USP9x/FAM acts
SMAD regulatory mechanisms involve two members of the deubiqui- to remove the ubiquitin moiety in the cytoplasm permitting SMAD4 to
tinating enzyme JAMM subfamily, associated molecule with the SH3 once again form a complex with phosphorylated forms of SMAD2. The
domain of STAM (AMSH) and its homologue AMSH-2 sequester SMAD6 juxtaposed activities of ECTO and USP9X function as a feedback loop to
and SMAD7, respectively, suppressing the inhibitory action of these I- regulate overall SMAD transcriptional output by initially permitting
SMADs towards their targets although it has never been conclusively SMAD2/SMAD4 complex formation and then by terminating tran-
determined if their deubiquitinating enzyme activity was required for scriptional output. Recent results have demonstrated the importance of
this function [95,96]. The exact targets of these DUBs are also unknown this as abrogation of ECTO expression impeded proper embryonic de-
but it is thought that AMSH is required for the regulation of TβR velopment [103]. Loss of SMAD4 is a common event in pancreatic
turnover by the endosomal sorting complexes required for transport cancer and colon cancer, and loss of USP9X has recently been shown to
(ESCRT) formation [97]. collaborate with KRAS to induce pancreatic cancer. Although the role of
R-SMADs are also regulated by DUBs. OTUB1 has been found to USP9X in this scenario may likely be due to the stabilization of YAP it
modulate TGFβ signalling by promoting the stabilization of only the cannot be discounted that inhibition of the TGFβ plays an integral role
phosphorylated form of SMAD2/3 [98]. Interestingly, it was found that in this process.
the mechanism is non-canonical and non-catalytic: instead of OTUB1
directly deubiquitinating SMAD2/3 rather it interacts with the E2 li-
3. The role of ubiquitin modifying enzymes in the hippo pathway
gases prevents the transfer of ubiquitin from the E2 ligase to the E3.
This non-canonical functionality of OTUB1 has been observed prior on
The importance of the Hippo pathway was original recognized in
multiple occasions [99–102]. Importantly, OTUB1's role in modulating
Drosophila through the use of genetic mosaic screens whereby random
TGFβ output has been established as a knockdown resulted in an in-
loss-of-function mutations in components of this pathway gave rise to
hibition of TGFβ induced migration [98] This lends support of a po-
severe organ overgrowth. It was later elucidated that these observed
tential role of OTUB1 in cancer, though this is yet to be verified.
effects were the result of increased cellular proliferation, decreased

463
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

apoptosis and enhanced progenitor stem cell renewal. The observation adaptor SAV1 concomitantly ubiquitinating and degrading both SAV1
that this pathway is also conserved in mammals has resulted in a flurry and LATS. Interestingly, the upstream kinase MST1 negatively regulates
of intensive research. The integrity of the Hippo pathway is absolutely the ability of NEDD4 to target SAV1 but in a disconnected fashion is
essential in early development. In some adult organs however, Hippo unable to inhibit NEDD4 mediated degradation of LATS [110]. Fur-
activity is expendable for normal homeostasis but is indispensable for thermore, the authors indicate that the proficiency of MST1 to inhibit
tissue repair and regeneration upon damage. Furthermore, experi- NEDD4 activity towards SAV1 is highly dependent on confluency of the
mental evidence has indicated a strong correlation between abnormal cells. In such a scenario high cell density activates the Hippo pathway
Hippo pathway activation and oncogenesis. The disparity between the by increasing phosphorylated active MST1 releasing the negative in-
requirement of Hippo signalling in cancer and its relative dispensability hibition of NEDD4 towards SAV1 and silencing YAP transcription. In-
in adult human tissues has led to the Hippo pathway being considered cidentally, of the NEDD4 family members only NEDD4 is capable of
as a very attractive therapeutic target in cancer. Several early phase binding SAV1 indicating that NEDD4, ITCH, and WWP1 interact with
clinical trials have been initiated and it will be interesting to see if any other adaptor proteins to regulate overall LATS1/2 levels. Similarly, in
clinical responses will be observed in patients receiving these com- response to hypoxia the E3 ubiquitin ligase SIAH2 ubiquitinates LATS2,
pounds. resulting in its destabilization [114]. Finally, the RING ligase PRAJA2
The canonical Hippo pathway consists of two central components, induces proteasome mediated degradation of MOB1 [115].
regulatory and transcriptional, which control overall Hippo activity. Mechanotransduction and the associated actin dynamics translate
The core regulatory component is a kinase cassette comprised of the the cells kinetic physical signals with the required downstream che-
Mammalian sterile-20-like (MST1/2, orthologue of Drosophila Hippo) mical cues to direct both short term and long term signalling. How actin
and the adaptor protein Salvador family WW domain-containing pro- specifically plays a role in downstream YAP and TAZ signalling remains
tein 1 (SAV1). Complex formation of MST1/2 and SAV1 permits to be elucidated. Angiomotin (AMOT) is a filamentous actin binding
phosphorylation and activation of large tumour suppressor 1(LATS1) protein that predominantly localizes to cellular junctions. AMOT and
and LATS2 and the LATS co-factor MOB kinase activator protein 1A two AMOT-like proteins, AMOTL1 and AMOTL2 regulate Hippo
(MOB1A) or MOB1B (reviewed in [104]). pathway activation by directly binding YAP and TAZ resulting in cy-
The transcriptional component of the pathway is composed of the toplasmic retention of both proteins [116]. Interestingly, the interac-
yes-associated protein (YAP) and its paralogue, transcriptional co-ac- tion between AMOT and YAP or TAZ occurs independent of YAP and
tivator with PDZ-binding motif (TAZ). These two transcription factors TAZ phosphorylation by LATS even though LATS enhances complex
shuttle back and forth between the cytoplasm and the nucleus where formation between these proteins by directly phosphorylating AMOT
they regulate transcription of a multitude of genes primarily through itself [117]. This indicates that LATS inhibits the nuclear localisation of
TEA domain family members (TEAD). When the Hippo pathway is ac- YAP and TAZ by two independent mechanisms; enhanced AMOT
tivated the LATS/MOB complex directly phosphorylates and inhibits complex formation and targeting YAP and TAZ for proteasomal medi-
the transcriptional co-activators YAP and TAZ through 14–3-3 mediated ated degradation. AMOT proteins themselves have been shown to be
cytoplasmic retention. The Hippo pathway is further regulated by nu- ubiquitinated on multiple residues with opposing effects on protein
merous upstream molecules, which regulate Hippo signalling through a function mediated by different ubiquitin topologies. Mono-ubiquitina-
multitude of post-translational modifications (reviewed extensively in tion of K347 and K408 is required for AMOTL2 to bind to the LATS1/2
[105–107]). Additionally, the mechanism of action of ubiquitination complex and may act as a scaffold to permit LATS phosphorylation and
and deubiquitination in the Hippo pathway is starting to fall into place inactivation of YAP and TAZ [118]. In contrast, polyubiquitination at
(Fig. 3). The most documented of these is the priming of YAP and TAZ K496 of AMOT targets AMOT for degradation resulting in YAP nuclear
for proteasomal mediated degradation following phosphorylation by localization [15]. Curiously however, both of these studies identify
LATS. LATS phosphorylation of YAP and TAZ serves to retain YAP and USP9X as the deubiquitinating enzyme regulating both the mono-
TAZ in the cytoplasm while concomitantly priming them for degrada- ubiquitination and polyubiquitination of AMOT. This contradictory
tion. The phosphorylation by LATS occurs in a unique phosphodegron result would suggest that deubiquitination of monoubiquitinated
site permitting recognition by the β-TrCP/SCF ubiquitin ligase complex. AMOTL2 by USP9X would inhibit the ability of LATS to phosphorylated
Recently, it has also been demonstrated that the RAS oncogene may YAP and TAZ, permitting YAP and TAZ to translocate to the nucleus and
serve to inhibit YAP protein turnover through two independent me- induce target gene expression. In contrast, however, Nguyen and col-
chanisms. RAS has been shown to downregulate the ubiquitin ligase leagues demonstrate that USP9X deubiquitinates polyubiquitinated
complex substrate recognition factors SOCS5/6 which serves to target AMOT stabilizing AMOT and sequestering YAP and TAZ in the cyto-
YAP to the B/C-Cullin 5 ubiquitin ligase for proteasomal degradation. plasm [15]. Further work will be needed to address the differences
Furthermore, the RAS isoform, H-RAS, induces the formation of MST1/ between these two studies and identify the function of USP9X in the
MST2 heterodimers muffling the kinase activity of MST1 towards LATS YAP pathway. Recently, several groups have shown the importance of a
resulting in downregulation of the pathway and YAP/TAZ nuclear lo- functional YAP1 pathway in KRAS mediated oncogenesis
calization [108]. [108,119–121]. In particular two independent studies revealed that loss
While degradation of YAP and TAZ is critical to the overall tumour of YAP1 expression prevented cancer progression in KRAS driven
suppressor activity of the Hippo pathway, ubiquitin mediated regula- mouse models of pancreatic ductal adenocarcinoma and lung cancer
tion of pathway has been demonstrated to act at multiple nodes along [122,123]. Data by Nguyen et al. indicate that USP9X transcript levels
its axis. A number of members of the Neural Precursor Cell Expressed, are significantly lower in thyroid, prostate, liver, hepatocellular carci-
Developmentally Down-Regulated 4 (NEDD4) family of E3 ubiquitin noma and kidney renal clear cell carcinoma with low USP9X expression
ligases including the E3 ubiquitin-protein ligase Itchy homologue correlating with significantly worse disease free survival in these later
(ITCH), WW Domain Containing E3 Ubiquitin Protein Ligase 1(WWP1), patients [15]. Also, USP9X appears to be a major tumour suppressor
and NEDD4 itself have been shown to regulate the stability and abun- gene in pancreatic ductal adenocarcinoma, as genetic abalation of
dance of LATS1 kinase [109–112]. Although these homologues share a USP9X co-operated with KRAS to accelerate pancreatic tumourigenesis
common functional architecture suggesting redundancy in their activity [16]. Furthermore, USP9X has been identified to mutated or deleted in
recent work has begun to identify the mechanisms that underlie their 4% of pancreatic adenocarcinomas. Ostensibly in the above pre-clinical
specificity [113]. As such their activity towards LATS may be likely contexts, loss of USP9X stabilizes YAP therefore indirectly enhancing
attributed to particular extracellular signals leading to specific complex RAS driven oncogenesis through YAP activation. As YAP inhibitors are
formation. An example of this has been shown to specifically occur with currently being tested in early phase clinical trials the potential that
NEDD4. NEDD4 directly binds to the previously mentioned LATS USP9X may be a biomarker for response for YAP inhibition must be

464
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

established. in contrast, TGF-α reduces tyrosine phosphorylation [131]. This phos-


The stability of AMOT does not only appear to be regulated by phorylation of USP8 at S680 permits complex formation with 14-3-3
USP9X, but recent data has also shown that the deubiquitinating en- proteins [131]. 14-3-3 proteins are highly important for regulation of
zyme DUB3 plays integral role in AMOT stability. However, like a various pathways in cells including cell cycle progression, DNA da-
number of DUBs in the TGFβ pathway DUB3 appears to act at several mage, apoptosis, transcriptional regulation of genes etc. It has been
levels to regulate Hippo pathway activation. First, DUB3 deubiquiti- postulated that 14-3-3 binding to USP8 may result in: alterations in its
nates and stabilizes the E3 ligase ITCH. As, ITCH mediated ubiquiti- enzymatic activity, changes in subcellular localisation or 14-3-3 may in
nation has been shown to promote the turnover of LATS and AMOT fact mediate the binding of other proteins affecting USP8 activity in an
proteins this suggests that DUB3 acts to enhance YAP activity. undefined way [132]. However, some studies have shown the role of
Interestingly DUB3 also interacts with and deubiquitinates AMOT, USP8 in deubiquitinating and stabilizing a RING finger containing E3
AMOTL1, and the kinases LATS1 and LATS2 to promote their stability Ubiquitin ligase called Nrdp1 which is involved in degradation of
and in so doing decrease YAP activity [124]. The authors hypothesis the ERBB3 and ERBB4 receptor tyrosine kinases [133]. These results are
primary function of DUB3 is to stabilize the scaffold protein AMOT opposite of the results seen by Niendorf et al. [130] showing that de-
permitting an unperturbed complex formation between AMOT and the letion of USP8 leads to lower level of ERBB3. Further investigation is
LATS kinases to downregulate YAP. The purpose of the seemingly needed to completely characterize the role of USP8 in regulation of
contradictory function of DUB3 with respect to ITCH in this setting ERBB receptor tyrosine kinase family. Possibly, the functional network
remains elusive but it is feasible that DUB3 binding to the AMOT/LATS between USP8, Nrdp1 and their targets such as EGFR family receptors is
complex may be regulated by post translational modifications whereby tissue and context specific dependent.
in the event that loss of binding to AMOT occurs, DUB3 can then act as Similarly, it has been shown that ubiquitin specific protease 2A
a potent activator of the pathway by stabilizing ITCH subsequently (USP2A) is localized in endosomes and prevents EGFR degradation
leading to degradation of AMOT and LATS. [134]. Liu et al. showed that overexpressing wild type USP2A, but not
mutant USP2A, led to stabilization of EGFR and enhanced downstream
4. The role of ubiquitin modifying enzymes in the MAPK pathway signalling. Furthermore, overexpressing USP2A in human bladder cells
significantly extended the duration of response to Heparin binding EGF
Mitogen-activated protein kinases (MAPKs) are protein Ser/Thr ki- like growth factor (HB-EGF) [135]. USP2A overexpressed cells could
nases that convert proliferative signals at the cell surface receptors into maintain high level of p-ERK from 5 to 60 min after treatment with HB-
a wide range of cellular responses. The importance of this pathway lies EGF while control cells showed only a transient increase in p-ERK level.
in maintaining a sensor dependent dynamic communication between This was probably due to the potential role of USP2A in deubiquiti-
cells and their surrounding environment. Many studies have empha- nating and stabilizing EGFR receptor.
sized sustained proliferative signalling due to hyperactive MAPK Insulin Growth Factor recptor-1 (IGF-1) has been shown as an im-
pathway as one of the main hallmarks of neoplastic diseases [125]. In portant initiator of MAPK and PI3K pathway activation. Intriguingly,
normal cells the classical MAPK cascade is sequentially relayed through mutant K1003R IGF-1R, which is not able to bind to ATP, is markedly
RAS induced phosphorlyation of MAPKKKs A-RAF, B-RAF, and C-RAF less ubiquitinated than wild type IGF-1R upon stimulation with IGF,
(also known as RAF1) following to MAPKKs MEK1 and MEK2, and suggesting that ubiquitination of IGF-1R is dependent on its autopho-
eventually MAPKs ERK1 and ERK2. The culmination of ERK signalling sphorylation [136]. Also, C-terminal domain of IGF-1R is needed for
results in regulation of genes involved in activation of cell cycle, pro- ubiquitination and IGF induced ERK activation. C-terminal truncated
liferation, survival, cell migration among others. In order to maintain a IGF-1R (Δ1245) is not ubiquitinated after IGF treatment and does not
systemic balance and to ensure that extracellular signals elicit appro- induce MAPK pathway activation while no change was reported in PI3K
priate downstream responses a number of mechanisms have evolved to pathway activation [136]. Moreover, treating 3T3-like fibroblasts with
regulate ERK activation, including ubiquitination (Fig. 4). The fact that lysosome inhibitor (Chloroquine) completely rescued IGF-1R protein
ubiquitination is reversible and that ubiquitinated proteins are known level from degradation while proteasome inhibitor (epoxomicine) was
to interact with a plethora of ubiquitin interacting proteins involved in only able to partly rescue this receptor [136]. Although, degradation of
signalling highlights the influence of ubiquitination as not only a de- truncated Δ1245 IGF-1R, which is not ubiquitinated, was completely
gradative signal. In no other context is this more evident than in the rescued by lysosome inhibitor, further studies are needed to investigate
crucial process of endocytosis. Initial activation of most pathways is whether ubiquitination of IGF-1R has any role in degradation of this
stimulated by ligand induced dimerization of cell surface receptors re- receptor.
sulting in autophosphorylation of Tyr residues in the intercellular do- Receptor Tyrosine Kinases (RTKs) such as EGFR and IGF-1R bind
main creating docking sites for protein adaptors. This autopho- growth factor receptor bound protein-2 (GRB2) following ligand in-
sphorylation event is more often than not associated with concomitant duced activation. GRB2 subsequently recruits the nucleotide exchange
ubiquitination of the receptors. The addition of the ubiquitin moiety is factor son of sevenless (SOS). The activity of downstream GTPase
required for cargo internalization and acts as a pilot shuttling the in- protein, RAS, depends on the ability of SOS to convert inactive RAS-
ternalized receptor towards active signalling nodes, recycling chutes, or GDP to the activate RAS-GTP. Activated RAS leads to phosphorylation
a degradative fate. As a number of excellent reviews on the role of and consequently sequential activation of the MAPKKK, MAPKK, and
ubiquitination in endocytosis has been written we will focus this seg- MAPKs. c-CBL is a RING finger E3 ligase involved in the regulation of
ment on the ubiquitination enzymes which effect canonical down- RTKs and contains three different domains in its N-terminal region in-
stream MAPK signalling [126,127]. Briefly, however, it must be noted cluding 4 helix bundle domains (4H), EF hand like domain (EF) and
that regulation of internalization and endosomal sorting involves the SH2 like domain. Together these three domains form what is known as
concerted action of an assembly of ligases and DUBs including for ex- a Tyrosine Kinase Binding (TKB) domain. The TKB supports the binding
ample USP8, USP2A, USP9X, AMSH, and Cezanne [126,128]. USP8 has of c-CBL to phosphotyrosine sites (Y1045 in EGFR) on receptor tyrosine
been reported as an important factor for rescuing transmembrane re- kinases such as EGFR, ERBB2 and ERBB4 [137,138]. c-CBL is also re-
ceptors such as EGFR, HGFR (c-MET), ERBB2 and ERBB3 receptors cruited to EGFR through an adaptor protein, GRB2 which binds to
from endocytosis mediated lysosomal degradation thereby further phosphorylated tyrosine sites on EGFR (mainly Y1068 and Y1086)
mediating downstream MAPK activation [129,130]. Interestingly, promoting ubiquitination and degradation of this receptor [137,138].
overexpression of constitutively active mutant SRC (SRC Y527A) sig- Upon complex formation with the RTK's c-CBL is itself phosphorylated
nificantly increases USP8 tyrosine phosphorylation [131]. Moreover, at tyrosine Y371 allowing the previously guarded E2 binding domain of
EGF stimulation of EGFR induces USP8 tyrosine phosphorylation while, c-CBL to become exposed and thus permitting full activation of the E3

465
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

Fig. 4. Schematic overview of regulation of MAPK pathway by Ubiquitin ligases and deubiquitinating enzymes. (A) Activation of the pathway through binding of ligand binding to
receptor tyrosine kinase leads to phosphorylation of tyrosine sites on intracellular components of the receptor creating docking sites for GRB2. Upon binding of GRB2 to the receptor, SOS
converts inactive GDP-RAS to active GTP-RAS. Active RAS induces RAF activation which leads to phosphorylation and activation of downstream MEK and ERK. Phosphorylated ERK
translocates to the nucleus and induces transcription. Sprouty can also bind to phosphorylated sites on receptor and competitively inhibits the binding of the negative regulator c-CBL and
rescues the receptor from degradation. Both USP2A and USP8 can also independently deubiquitinate and stabilize the receptor. Activation of the pathway is maintained by through
SMURF2 and USP15. SMURF2 ubiquitinates and degrades the RAS negative regulator β-TrCP while USP15 deubiquitinates and stabilizes RAF. (B) The MAPK pathway is negatively
regulated through a number of different ubiquitin mediated mechanisms. Upon binding of ligand and receptor phosphorylation the E3 ligases c-CBL and Nrdp1 ubiquitinate the receptor
targeting the receptor for degradation. SPROUTY blocks the interaction of SOS and RAS and SPROUTY's interaction to RAF-1 blocks its kinase activity. β-TrCP ubiquitinates and degrades
RAS. Rabex-5 monoubiquitinates H-RAS and N-RAS and prevents their endosomal recycling into the membrane. IMP is a known ubiquitin ligase which targets the scaffold protein KSR
leading to its degradation. IMP autoubiquitination is reversed by USP15 stabilizing the protein. Several ubiquitin ligases and chaperones are responsible for degradation of RAF including
RNF149, HUWE1, XIAP and HSP90. Also, MEKK1 ligase is known to add ubiquitin chains to ERK and degrade this protein in proteasome dependent manner. The overall outcome of these
events is downregulation of pathway and inhibition of ERK mediated transcription.

ligase activity of c-CBL towards its bound RTK leading to proteosomal previously been described as having both overlapping and non-re-
mediated degradation of the RTKs [139]. dundant functions. Under physiological conditions RAS proteins con-
The adaptor protein SPROUTY has been commonly regarded as a tinuously cycle between an active GTP-bound state and inactive GDP-
negative regulator of MAPK pathway though negative regulation ap- bound form which allows RAS proteins to bind and activate its various
pears contextual; dependant on the specific RTK activated ligand. Like downstream substrates. The majority of RAS mutations typically affect
c-CBL SPROUTY contains a TKB binding motif with an affinity for hotspots within codons 12 and 13 most notably G12 V for KRAS. On a
phosphorylated EGFR tyrosine Y55 and in the event of EGF stimulation molecular level these mutations impair GTP-GDP hydrolysis which in
SPROUTY is able to bind instead of c-CBL inhibiting c-CBL ubiquiti- turn leads to permanent activation of RAS and persistent stimulation of
nation of EGFR [140]. Therefore, SPROUTY acts as a competitive in- downstream signalling pathways. The abundance and subcellular lo-
hibitor preventing c-CBL interaction with EGFR hence enhancing MAPK calisation of all isoforms of RAS are regulated by ubiquitination. In light
signalling pathway [141]. In contrast, SPROUTY tends to act as a ne- of this, though attempts to effectively pharmacologically inhibit K-RAS
gative regulator following exposure to fibroblast growth factor (FGF), have been unsuccessful, recent data suggests that targeted inhibition of
vascular-endothelial growth factor (VEGF), platelet- derived growth ubiquitin modifying enzymes may lead to downregulation of K-RAS
factor (PDGF), hepatocyte growth factor (HGF), glial-derived growth function. The F-box protein β-transducin repeat–containing protein (β-
factor (GDGF), or nerve growth factor [142]. It does so by acting as a TrCP) mediates polyubiquitination of all RAS isoforms, leading to
decoy adaptor for GRB2 preventing activation of RAS by inhibiting proteasome-dependent degradation of RAS [148]. Interestingly, this
interaction of GRB2 with RAS [143]. Secondly, SPROUTY can bind to process is partly mediated by Wnt/β-catenin signalling pathway
the cysteine rich domain to C-RAF kinase and blocking its kinase ac- whereby Axin and Adenomatous Polyposis Coli (APC) enhance the
tivity [144,145]. Interestingly, SPROUTY expression is induced by binding of the WD40 domain of β-TrCP with H-RAS leading to down-
MAPK pathway activation eluding to the existence of a negative feed- regulation of the transforming capability of H-RAS. These results em-
back loop [144]. phasize regulation of the RAS proteins by the WNT pathway.
In about one third of all human cancers one of three isoforms of RAS β-TrCP itself undergoes ubiquitin mediated degradation. The E3
(H-RAS, N-RAS and K-RAS) are mutated asserting the critical im- ligase SMURF2 monoubiquitinates its cognate ubiquitin-conjugating
portance of these small GTPases in proliferation and cell survival [146]. enzyme (E2), UBCH5/UBE2D, leading to the active E3:E2 complex
The primary difference in three isoforms of RAS lies in their subcellular formation resulting in the polyubiquitination and degradation of β-
localization which is mediated by a 25 amino acid C-terminus hy- TrCP [149]. Thus SMURF2 activity leads to the stabilization of K-RAS.
pervariable region (HVR) containing sites for farnesylation and palmi- Conversely, loss of SMURF2 expression by genetic means stabilized β-
toyilation for H-RAS and N-RAS and only farnesylation site (in the case TrCP and degraded K-RAS. Interestingly, the authors also observed that
of K-RAS) [147]. H-, N- and K-RAS, are ubiquitously expressed and have K-RAS mediated ubiquitination occurred at a higher level in mutant

466
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

forms of the protein, probably owing to its persistent GTP activation. inhibitor of apoptosis (ML-IAP) using siRNA leads to stabilization of C-
This data would suggest that effective targeting of SMURF2 may be a RAF and promotes migration [157,158]. Although, IAP family members
unique strategy to degrade mutant K-RAS in cancer cells. However, one contains a RING E3 Ubiquitin ligase domain, XIAP and c-IAP depletion
must consider that SMURF2 acts as a negative regulator of TGF-β sig- stabilized C-RAF level (≈2fold) in a RING domain independent manner
nalling and therefore targeting of SMURF2 may activate the pro-onco- [157]. Moreover, the same study showed the role of chaperons and co-
genic effects of TGF-β while simultaneously deregulating K-RAS. chaperons (mainly CHIP) in ubiquitination and degradation of C-RAF.
The endosomally localized Rabex5 (also known as RabGEF1) func- Carboxy terminal Hsc70 interacting protein, CHIP, is one of the im-
tions partly as an E3 ubiquitin ligase and promotes mono- and di-ubi- portant co-chaperons which conjugates E2 ubiquitin ligases to C-RAF.
quitination of H-RAS and N-RAS, leading to the anchoring of RAS at the However, interaction of C-RAF and CHIP requires XIAP as an adaptor
endosomes and reducing downstream signalling [150,151]. It has been protein which explains the increased ubiquitination and degradation of
shown that Rabex-5 contains an A20 like Zing finger ubiquitin ligase C-RAF by overexpression of the mutant RING domain deleted XIAP
domain (ZnF) which mediates the interaction of Rabex-5 with H-RAS [157].
and N-RAS. Interestingly, mutant H-RAS which is not able to be ubi- Similar to C-RAF, A-RAF and mutated B-RAF V600E (but not wild-
quitinated is significantly more effective than wild type H-RAS in its type B-RAF) are stabilized through HSP90 chaperone machinery [159].
activation of MAPK pathway highlighting the role of non-degradative HSP90 is required for C-RAF stability and membrane localization.
ubiquitination of RAS proteins in downregulation of MAPK pathway Geldanamycin, a HSP90 binding antibiotic, disrupts complex formation
[147]. The overall fate of endosomal bound RAS remains un- of C-RAF and HSP90 and induces ubiquitin dependent degradation of C-
determined, most likely is it either targeted for endosomal mediated RAF [160]. C-RAF mediated downstream signalling was inhibited by
recycling or shunted towards the proteasomal degradation pathway. Geldanamycine possibly through reduction of C-RAF level. However,
Furthermore, reversal of this ubiquitination at this location by an en- the signalling and C-RAF level was maintained in presence of protea-
dosomal localized deubiquitinating enzyme has not yet been described. some inhibitors, even in presence of Glendamycine [161]. Also, mutant
Individual RAS family members can also undergo monoubiquitina- B-RAF V600E, which is frequently mutated in different types of cancer
tion at K117 and K147. It has been proposed that targeted ubiquitina- including melanoma, colorectal and thyroid cancer, is a HSP90 client
tion of K-RAS specifically at K147 impairs regulator-mediated GTP [159]. Although, HSP90-cdc37 complex was involved in degradation of
hydrolysis leading to markedly increased activation of MAPK pathway. different B-RAF mutants (V600E, V600D, G465V, G468A), there was no
In contrast, monoubiquitination of H-RAS at K117 accelerates intrinsic change in level of wild type B-RAF after 17-AGG (HSP90 inhibitor)
nucleotide exchange promoting GTP loading [152]. Interestingly, these treatment, consistently shown in different melanoma cell lines [159].
hyperactivated forms of RAS demonstrate enhanced interaction binding Interestingly, RNF149 was reported as an E3 ligase responsible for
with downstream effectors and overall activation of the pathway degradation of wild type B-RAF, but not mutant B-RAF [162]. siRNA
through monoubiquitination at distinct sites eliciting distinct mechan- mediated downregulation of RNF149 leads to stabilization of wild type
isms of action. Though the identity of both the E3 ligase and the B-RAF and simvastatin treatment which induces RNF149 expression
counteracting DUB that regulates this site remains elusive, it is leads to lower level of wildtype B-RAF, without altering mutant V600E
tempting to speculate that a balance exists between E3 ligase activity B-RAF level [162]. Interestingly, some studies have investigated the
and DUB activity to regulate overall RAS activity through stabilization role of RAF proteins and dysregulation of its HSP90 dependent de-
and localization of the protein. gradation in the non-cancer context of metabolic diseases such as dia-
Impedes mitogenic signal propagation (IMP) is a Ring finger E3 li- betes [163]. It has been shown that, methylglyoxal, a physiological
gase which negatively regulates KSR and inhibits MAPK pathway ac- metabolite of glucose which is accumulated in serum of diabetic pa-
tivation through the prevention of RAF and MEK complex formation. tients, significantly reduces C-RAF level through ubiquitin dependent
Binding of extracellular ligands to RTKs and activation of RAS leads to degradation [163].
activation of IMP E3 ligase activity and its autoubiquitination and Extracellular signal related kinases (ERKs) are a family of proteins
subsequent degradation. Therefore, increased IMP autoubiquitination which are responsible for transmitting the extracellular signal into
following RAS activation leads to KSR stabilization and higher level of nucleus through phosphorylation or regulation of various transcription
phosphorylated ERK1/2 [153,154]. How activation of RAS causes in- factors including ETS, ELK, MNK, RSK etc. ERK family kinases are
creased E3 ligase activity of IMP remains an unanswered question classified into two categories depending on scaffold dependence for
which requires further investigation. Recently, some studies have re- activation and function. C-terminal domain containing MAPKs such as
ported the role of USP15 in deubiquitinating and rescuing IMP from ERK3 and ERK7/8 can exist in individual subunits independent of
degradation [155]. Although, USP15 and USP4 both are involved in scaffold binding. The second category includes the classical ERK family
deubiquitination of IMP, depletion of only USP15, not USP4, destabi- members ERK1 and ERK2. The signalling axis of this pathway requires
lizes IMP promoting its proteasomal dependent degradation [155]. scaffolding proteins for their proper activation. As an example KSR
Hayes et al. showed that the interaction of USP15 and IMP is mediated (Kinase suppressor of RAS) is needed as a scaffold protein to pre-
through DUSP-UBL domain of USP15 and coiled coil region of IMP. assemble and engage the multiple components of the MAPK cascade for
Moreover, the same study showed the role of USP15 in stabilizing C- efficient ERK1/2 activation [153].
RAF [155]. Although, USP15 depletion reduced the level of C-RAF, it ERKs have been shown to be regulated by ubiquitination/deubi-
had no effect on the level of B-RAF and p-MEK in B-RAFV600E har- quitination system. Upstream of ERK, MEK kinase1 (MEKK1) is able to
bouring melanoma cells [155]. activate MEKs in both JNK and ERK1/2 pathways. PHD (plant home-
HUWE1 a HECT family E3 ligase has recently been demonstrated to odomain) domain of MEKK1 which is a Ring finger like domain shows
regulate of C-RAF protein level, but not B-RAF and A-RAF levels [156]. E3 ubiquitin ligase activity and is responsible for ubiquitination and
HUWE1 binds indirectly to C-RAF through a scaffold protein called degradation of ERK1/2 [164]. Therefore, MEKK1 activates and down-
SHOC2 and ubiquitinates both SHOC2 and C-RAF proteins. As ex- regulates ERK1/2 through its kinase and PHD domain, respectively
pected, depletion of SHOC2 abrogates HUWE1 mediated C-RAF ubi- [164]. The PHD domain in MEKK1 contains 7 cysteines and a histidine
quitination and degradation [156]. Downregulation HUWE1 expression which resembles the structure of other known E3 ubiquitin ligases. It
by shRNA stabilized C-RAF and SHOC2 and increased p-ERK levels in has also been reported that the PHD domain is found in many proteins
different cells lines, demonstrating HUWE1 as a negative regulator of which are involved in chromatin mediated transcriptional regulation
MAPK signalling. [164]. ERK1/2 maintains homeostasis by inhibiting apoptosis. Treat-
Some studies have shown that silencing X linked Inhibitor of ment with sorbitol results in cellular hyperosmotic conditions. This
apoptosis (XIAP), cellular inhibitor of apoptosis (c-IAP) or melanoma increases the interaction of MEKK1 and ERK1/2 leading to

467
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

downregulation of ERK1/2 through proteasome mediated degradation highlights the complex role of ubiquitination towards its targeted
mechanism allowing for apoptosis to occur [164]. This demonstrates substrates and may suggest a tissue specific regulatory function of p85
the crucial nature of ubiquitination along the ERK1/2 axis in main- towards PI3K activation.
taining homeostasis at the organism level. Like p85, IRS-1 is itself a target of proteasomal mediated degrada-
tion. CUL7 E3 ligase is a component of the SCF complex including
5. The role of ubiquitin modifying enzymes in the PI3K pathway FBW8, SKP1, and the ROC RING finger protein. As part of negative
feedback loop S6K activation downstream of mTOR phosphorylates
P13K-AKT-mTOR (PAM) pathway is a critical signalling pathway IRS-1 at S307 stimulating the interaction of IRS-1 with the substrate
involved in cellular growth and survival. Genetic aberrations that lead recognition subunit FBW8 and recruiting the SCF complex consequently
to constitutive activation of the pathway are frequently observed in resulting in IRS-1 ubiquitination and degradation. Treatment with the
human cancers. However, due its aberrant behavior and irregular cross- mTOR inhibitor rapamycin repressed IRS-1 phosphorylation and in-
talk with other signalling pathways targeted therapy with single agent hibited the CUL7 E3 ligase mediated degradation of IRS-1 revealing the
PI3K pathway inhibitors has generated disappointing results clinically. importance of mTOR/S6 activities in ubiquitin mediated upstream
Renewed enthusiasm analyzing various combination therapies, in- regulation of IRS-1 by the SCF complex [171]. Likewise, SOCS con-
cluding strategies targeting CDK4/6 with PI3K inhibitors has revived taining E3 ligases has been reported to block insulin signalling by in-
hope that effective downregulation of the pathway will lead to pro- ducing ubiquitin mediated degradation of IRS1 and IRS2 and have been
longed responses in patients. Nevertheless, a better understanding of implicated in the mechanism for inflammation-induced insulin re-
the PI3K pathway is essential for this effort. sistance [172]. Several other ubiquitin ligases suppress IGF/insulin
PI3Ks as the major downstream effector of receptor tyrosine kinases signalling by inducing ubiquitin mediated proteasomal degradation of
(RTKs) and G protein coupled receptors (GPCRs), leads to the formation IRS-1/2 [173,174] whereas mono-ubiquitination of IRS-2 at single or
of a potent second messenger, PIP3 (Phosphatidylinositol (3,4,5)-tri- multiple sites by NEDD4 has been reported to enhance IGF signalling in
phosphate) which transduces the signals from various growth factors a manner dependent on the ubiquitin binding protein Epsin1 [175].
and cytokines and assists in AKT activation. The serine threonine kinase Ubiquitination of IRS proteins has been shown to be reversed by deu-
AKT once activated plays a vital role in the regulation of cell survival in biquitinating enzyme USP7. USP7 forms a complex with IRS1/2 deu-
a variety of human neoplastic diseases and polices a range of cellular biquitinating and stabilizing IRS1/2. Interestingly, complex formation
processes, including cell survival, cell cycle progression, cytoskeletal between IRS and USP7 is modulated by PI3K signalling as the addition
organization, vesicle trafficking, glucose transport, and platelet func- of insulin led to the dissociation between USP7 and IRS1/2 enhancing
tion. Along with a number of activating mutations within components IRS1/2 degradation [176]. This dissociation was prevented by treat-
of the PAM pathway a number of inhibitory mutations have also been ment with the pan PI3K inhibitor LY294002. This suggests that IRS1/2
identified in the tumour suppressor phosphatase and tensin homologue mediated activation of the PI3K pathway following insulin exposure
deleted from chromosome 10 (PTEN). PTEN is a dual specific protein induces a dual feedback mechanism targeting IRS1/2 stability. Fol-
and lipid phosphatase whose primary function is to degrade the phos- lowing activation of the PI3K pathway S6K phosphorylates IRS1/2 re-
phoinositide products of PI3K hydrolyzing (3,4,5)-trisphosphate (PIP3) cruiting the SCF ligase complex targeting IRS1/2 for ubiquitination.
to phosphatidylinositol (4,5)-bisphosphate (PIP2) [165,166]. In terms Concomitantly, PI3K activation dissociates USP7 from the IRS1/2
of enzymatic activity of the various component of PI3K-AKT-mTOR complex ensuring a rapid downregulation of activated IRS1/2.
pathway including AKT and PTEN the role of phosphorylation has been
well established however, very little is known about the role ubiquitin 5.2. Ubiquitin mediated regulation of AKT
modifying enzymes in this process (Fig. 5).
Substrate ubiquitination and targeting of proteins to the proteasome
5.1. Ubiquitin mediated regulation of PI3K and IRS is a dynamic process and can have a great impact on progression of
oncogenesis. Recent studies suggest that ubiquitination is equally im-
Phosphatidylinositol 3-kinase (PI3K) is a conserved hetero-dimer portant as phosphorylation for activation of PAM pathway components
intracellular lipid kinase consisting of an 85-kD regulatory subunit and as such it is unsurprising that pharmaceutical inhibition of a
bound to a 110-kD catalytic subunit. The phosphoinositol-3-kinase fa- number of ubiquitin modifying enzymes with respect to PI3K activation
mily is grouped into three different classes: Class I, Class II and Class III. is being explored, including a number of enzymes targeting AKT. AKT is
In response to various growth factors class IA PI3Ks get activated either a member of the AGC family of kinases, the members of which possess a
through direct binding with the activated receptors or through adapter pleckstrin homology (PH) domain at its N-terminus. As indicated above
molecules such as phosphotyrosyl insulin receptor substrate (IRS). The PI3K phosphorylates the inositol ring of PI(4,5)P2 at the D-3 position to
p85 subunit of PI3K has been shown to be regulated by several E3 li- form PI(3,4,5)P3, which is required for the binding of the PH-domain of
gases including SCF-FBXL2, CHIP, and CBL. SCF-FBXL2 catalyses ubi- AKT to the plasma membrane. This permits AKT to be phosphorylated
quitination and hence degradation of monomeric p85 [167]. Curiously, by phosphoinositol-dependent kinase 1 (PDK1) within its catalytic do-
FBXL2 mediated degradation of p85 appears to enhance PI3K activation main at T308. Following pathway activation AKT is subsequently
rather the inhibit it. The authors rationalize that FBXL2 preferentially phosphorylated at S473 by ribosome-associated RICTOR-mTOR com-
targets excess free monomeric p85 thereby limiting the ability of the plex (mTORC2). Phosphorylation of both T308 and S473 is required for
free p85 to outcompete the p85-p110 heterodimer for binding to IRS1 maximum kinase activity of AKT. Similarly, AKT kinase activity can be
[168]. Recently, it has been demonstrated that the ErbB3-binding both regulated by K63-linked ubiquitination and K48 linked ubiquiti-
protein 1 (EBP1) couples p85 to the HSP70/CHIP complex. The cha- nation. TRAF6, TRAF4, NEDD4, and SKP2 all regulate AKT signalling
perone-dependent ubiquitin ligase (CHIP) drives p85 ubiquitination by promoting K63 linked ubiquitination. K63-linked ubiquitination has
and ultimately proteasomal mediated degradation of p85 [169]. It has been revealed to be critical for activation of AKT by enabling its
been also reported that CBL family ubiquitin ligases not only mediates membrane recruitment. TRAF6 ubiquitinates AKT at two lysines (K8
polyubiquitination of p85 in T cells but under certain conditions can and K14) within its PH domain promoting translocation of AKT to
regulate p85 compartmentalization and protein-protein interactions at plasma membrane and enhancing AKT T308 phosphorylation and ac-
the cell membrane [170]. CBL-B recruits p85 to CD28 and T cell antigen tivation [177]. Curiously, K63 ubiquitination at these two residues is
receptor and negatively regulates p85 in a proteolysis-independent not required for PIP3 binding even though the K14 residue resides
manner. The contrasting effects of ubiquitin mediated degradation and within the PIP3 binding pocket. It has therefore been speculated that
regulation of p85 on downstream PI3K activity in these three studies ubiquitination may enhance a conformational change within the

468
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

Fig. 5. Schematic overview of regulation of PI3K pathway by Ubiquitin ligases and deubiquitinating enzymes. (A). Ubiquitin mediated downregulation of the PI3K pathway. PI3Ks are
downstream effectors of receptor tyrosine kinases (RTKs) and G protein coupled receptors (GPCRs). Phosphatidylinositol 3-kinase (PI3K) is a hetero-dimer conserved intracellular lipid
kinase consisting of an 85-KD regulatory subunit bound to a 110-KD catalytic subunit. CHIP and CBL regulate stability and function of p85 respectively through ubiquitination. Like p85,
the RTK interacting protein IRS undergoes ubiquitination mediated degradation through CUL7 and SOCS. The downstream serine/threonine kinase AKT has been shown to undergo K48
ubiquitinated by the ligases MULAN, TTC3, BRCA1 and CHIP. These ligases degrade AKT downregulating PI3K pathway activity. Likewise, the DUB CYLD prevents activation of AKT by
counteracting K63 linked ubiquitination and inhibiting AKT recruitment to membrane. Furthermore, the phosphatase PHLPP1 functions in complex with FKBP51 to dephosphorylate and
inhibit AKT. USP49 stabilizes FKBP1 further enhancing AKT pathway inhibition by promoting interaction of PHLPP1 to AKT. Cul4-DDB1-Fbw5 mediates TSC2 stability and inhibiting
TSC1/TSC2 complex turnover resulting in constitutive downregulation of the mTOR activator, RHEB. mTOR is the key component of mTORC1 (mTOR, mLST8, RAPTOR) and mTORC2
(mTOR, mLST8, RICTOR) whereby mTORC1 enhances phosphorylation of downstream S6K1 and eIF4E while mTORC2 acts through a positive feedback loop to phosphorylate AKT at
S473 leading to full activation of AKT. Cul1-Skp-Fbw7 and USP9X negatively regulate mTOR in ubiquitin dependent manner, however, the manner through which USP9X regulates mTOR
stability is unknown. The mTORC2 component RICTOR has been shown to be ubiquitinated and degraded by SCF-FBXL7. Interestingly mROC1 and mTORC2 complex formation is
regulated by a number of ubiquitin modifying enzymes. mLST8, another component of both mTORC complexes, undergoes K63 mediated ubiquitination by TRAF2 hindering mTORC2
formation while increasing mTORC1. A mechanism offset by the DUB OTUD7B. In a similar fashion CUL4-DDB1 ubiquitinates RAPTOR limiting mTORC1 formation but enhancing
mTORC2 formation. A mechanism reversed in this case by UCH-L1. The tumour suppressor PTEN negatively regulates PI3K pathway by inhibiting formation of PIP3 but also plays an
undefined role as a tumour suppressor in the nucleus. PTEN also undergo various post translational modification including ubiquitination. PTEN was shown to be monoubiquitinated by
NEDD4-1 enhancing its nuclear translocation. Furthermore, two DUBs USP13 and OTUD3 deubiquitinate and stabilize PTEN thereby aiding in its tumour suppressor activities. (B)
Ubiquitin mediated upregulation of the PI3K pathway. The E3 ligase FBXL2 mediates degradation of p85 but interestingly this degradation enhances PI3K activity by preferentially
targeting excess free monomeric p85 thereby limiting the ability of free p85 to outcompete the p85-p1110 heterodimer for binding to IRS1. NEDD4 has been reported to enhance IGF
signalling by monoubiquitinating IRS at single or multiple sites. Similarly, USP7 counteracts ubiquitination of and stabilizes IRS1/2. PDK1 which phosphorylates and activates AKT is
monoubiquitinated and undergoes deubiquitinated by USP4. Although the exact function of this DUB with respect to PDK1 needs to be explored. Membrane recruitment of AKT is a
critical step for its activation. Various ligases such as TRAF6, SKP2, NEDD4 and TRAF4 all mediate K63 linked ubiquitination of AKT recruiting AKT to the plasma membrane. The E3
ligase SCF-β-TrCP ubiquitinates and degrades the AKT negative regulator PHLPP1. Similarly, UCH-L1 which is a deubiquitinating enzyme suppress the level of PHLPP1 thus enhancing
AKT pathway. Deptor, thought to be a negative regulator of mTORC activity undergoes ubiquitination and degradation by the E3 ligase β-TrCP. S6K1, which is downstream of mTORC1
also undergoes ubiquitin mediated modification by ROC1. ROC1 might be responsible for turnover of S6K protein. PTEN itself is also negatively regulate by a number of ligases including
WWP1, XIAP, CHIP, NEDD4-1 targeting PTEN polyubiquitination and degrading PTEN. RFP also polyubiquitinates PTEN but rather then inducing degradation it functions by inhibiting
its activity. USP7 as a deubiquitinating enzyme counteract monoubiquitination of PTEN and causes its nuclear exclusion.

protein permitting complex formation with a ubiquitin binding factor degradation, the SCF complex (SKP2, CUL, FBOX), mediates K63 ubi-
resulting in the relocalisation of AKT from the cytosol to the plasma quitination of AKT [180]. For the most part SCF complexes promote
membrane. Interestingly, AKT mediated ubiquitination by TRAF6 is protein degradation through the transfer of K48 ubiquitin chains. Why
enhanced following growth factor stimuli. In fact, it has been noted that in this case SKP2-SCF complex promotes a non-proteolytic K63 linked
AKT K63 ubiquitination is mediated by a diverse set of E3 ligases de- ubiquitination of AKT remains a bit of mystery and this issue, un-
pending on the stimulus, with both TRAF6 and the neural precursor cell doubtedly, will need to be probed into further. It is important to note
expressed developmentally down-regulated protein 4 (NEDD4) having that all three of these ligases have been reported to promote cancer
been demonstrated to regulate AKT activity through K63 linked ubi- progression. Activating mutations in AKT itself are rarely observed,
quitination following IGF-1 signalling whereas SKP2 and TRAF4 func- with the exception of a single point mutation E17K within the PH do-
tion downstream of ErbB receptors [178,179]. Curiously, unlike IRS1/2 main [181]. Interestingly, this cancer associated mutant displays hy-
where the SCF complex targets IRS1/2 for proteosomal mediated perubiquitination of AKT, increased PIP3 binding and increased

469
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

membrane localization indicative of the importance of PH domain component in two distinct signalling complexes mTORC1 and mTORC2.
ubiquitination and AKT activation. mTORC1 is comprised of mTOR, the regulatory associated protein of
Counteracting these ligases the deubiquitinating enzyme CYLD acts mTOR (RAPTOR), mammalian lethal with Sec13 protein 8 (mLST8), the
as a tumour suppressor and negative regulator of AKT activation by proline rich AKT substrate 40 kDA (PRAS40), and DEP-domain-con-
apparently directly deubiquitinating K63 linked ubiquitin chains within taining mTOR-interacting protein (DEPTOR). mTORC2 comprises of six
the PH domain, terminating AKT activation [182,183]. Similar results different proteins including rapamycin-insensitive companion of mTOR
were observed in TRAF6 depleted cells. As it has previously been de- (RICTOR), mammalian stress-activated protein kinase interacting pro-
monstrated that CYLD deubiquitinates TRAF6 these results suggests tein (mSIN1), protein observed with RICTOR-1 (Protor-1), mLST8, and
that CYLD deubiquitinates and suppresses the activity of both the E3 DEPTOR [194]. Due to the importance of mTORC1/2 in cell growth and
ligase and its kindred substrate [184]. An effect which has been fre- metabolism it is unsurprising that deregulation of mTOR signalling is
quently observed [88,185]. Interestingly, K14 but not K8 was the cri- associated with various diseases some of which appear to be determined
tical residue for CYLD mediated deubiquitination. by aberrant ubiquitination of mTOR components. Even though both
Proteasomal mediated degradation of AKT is regulated by a plethora mTORC1 and mTORC2 contain a number of common components ac-
of ubiquitin ligases including tetratricopeptide repeat domain 3 (TTC3), tivation of mTORC1 and mTORC2 occurs through unique stimuli in-
breast cancer susceptibility gene 1 (BRCA1), chaperon-associated ubi- cluding growth factors or nutrients and PI3K signalling, respectively.
quitin ligase (CHIP), and mitochondrial ubiquitin ligase activator of NF- Correspondingly, mTORC1 and mTORC2 appear to be regulated
κB (MULAN) [186–189]. In the majority of these cases phosphorylation through both analogous and distinctive ubiquitination schemes.
and activation of AKT is a prerequisite determinant for ligase binding, mTORC1 primarily functions downstream of AKT to regulate pro-
AKT K48 ubiquitination, and subsequent degradation. TTC3 has been tein translation and cell growth by phosphorylating S6K1 and EIF4E-
shown to preferentially bind to phosphorylated forms of AKT. Inter- BP1 Eukaryotic translation initiation factor 4E-binding protein 1
estingly, as part of a self-regulation mechanism, AKT phosphorylates (EIF4E-BP1) [195]. It has been reported that DEPTOR is a naturally
TTC3 at S378, a site elucidated to be required for TTC3 activity as occurring inhibitor of both mTORC1 and mTORC2 [196]. DEPTOR is
mutation of TTC3 inhibited TTC3 mediated ubiquitination and de- targeted for ubiquitin mediated degradation by the SCF (Skp1-Cullin-F
gradation of AKT. box proteins)- βTrCP E3 ligase in response to serum stimulation,
The tumour suppressor BRCA1 is frequently mutated in breast and thereby promoting mTOR activation and cellular proliferation
ovarian cancers. BRCA1 activity regulates a number of signalling [197–199]. Similarly, RAPTOR and mLST8 interact with CUL4-DDB1, a
pathways involved in genome stability including DNA damage repair, core component of CUL4-DDB1-WDR ubiquitin E3 ligase complex, re-
cell cycle check point control, chromatin remodeling, transcriptional sulting in enhanced mTOR mediated signalling. Loss of CUL4B and
regulation, apoptosis, and ubiquitination. Like TTC3, BRCA1 is re- DDB1 diminished mTORC1-dependent phosphorylation of S6K and 4E-
cruited to phosphorylated isoforms of AKT and targets active AKT in- BP1 [200]. Interestingly, CUL4-DDB1 function does not appear to reg-
ducing K48 ubiquitin chain formation and degradation of the later ulate RAPTOR or mLST8 stability directly but functions to enhance
[186]. This work highlights the ubiquitin mediated tumour suppressive mTORC1 assembly. Counteracting this process ubiquitination of
function of BRCA1 in tumorigenesis. Similarly, the mitochondrial as- RAPTOR by CUL4-DDB1 is offset by the deubiquitinating enzyme UCH-
sociated ubiquitin ligase MULAN and CHIP target phosphorylated AKT. L1 [201]. In either setting a direct consequence of either CUL4B-DDB1
Interestingly, K48 ubiquitination of phosphorylated AKT can occur both or UCH-L1 function is the reorganization and assembly of competing
in the nucleus (BRCA1, TTC3) and in the cytoplasm (MULAN, CHIP) mTOR complexes. CUL4B-DDB1 supports mTORC1 complex formation
eluding to the potential role of AKT substrates in negative feedback while limiting mTORC2 while UCH-LI destabilizes mTORC1 enhancing
loops in the recruitment of these ligases in both the nucleus and cyto- mTORC2 assembly. Comparably, the E3 ligase TRAF2 has recently been
plasm to downregulate active AKT. It also suggests that to prevent shown to promote K63 mediated ubiquitination of mLST8 hindering
nuclear exclusion of AKT through its degradation in the cytoplasm by complex formation with the mTORC2 specific component SIN1 thereby
ubiquitin ligases a small proportion of phosphorylated AKT may be promoting mTORC1 formation, a process reversed by the deubiquiti-
specifically recruited to the nucleus to perform its nuclear functions. It nating enzyme OTUD7B (also known as CEZANNE) [202]. As such it
will be interesting to determine if the hyperactivated form of would be expected that both CUL4B-DDB1 and TRAF2 enhance
AKT(E17K) functions primarily in the cytoplasm or the nucleus and mTORC1 dependent S6K and 4E-BP1 phosphorylation while UCH-L1
ubiquitin mediated degradation of AKT (E17K) is altered at either of and OTUD7B enhance mTORC2 dependent AKT phosphorylation at
these locales. S473.
mTOR signalling can further be downregulated through a number of
5.3. Ubiquitin mediated regulation of PDK1 and mTOR ubiquitin dependent mechanisms with mTOR itself being negatively
regulated by both Cul1-Skp-Fbw7 E3 ligase and the DUB USP9X
AKT is phosphorylated at T308 by PDK1 and subsequently at S473 [203,204]. The targeting of the Cul1-Skp-Fbw7 complex to mTOR re-
by mTORC2 resulting in full activation of the protein. PDK1 (3- sults in the latter's ubiquitination and degradation while the mechanism
phospho-inositide-dependent kinase 1) is a serine/threonine kinase through which USP9X inhibits mTOR signalling remains ill defined. In
which phosphorylates and activates approximately twenty-three of the contrast, Cul4-DDB1-Fbw5 E3 ligase regulates TSC1/TSC2 complex
AGC family of protein kinases including the proto-oncogene AKT, turnover increasing in the constitutive activation of the downstream
Protein Kinase C (PKC) and the p70 S6 kinase (S6K) [190–192]. PDK1 mTOR activator RHEB. Downstream of mTOR ribosomal protein S6
plays critical role in cell proliferation and metabolism. Mono-ubiquiti- kinase has been demonstrated to be ubiquitinated and targeted for
nated isoforms of PDK1 have been reported in a various human cell degradation by the ligase ROC1 resulting in the inhibition of S6K ac-
lines suggesting PDK1 ubiquitination is a common process. The deu- tivity [205].
biquitinating enzyme USP4 co-localizes with PDK1 at the plasma- In addition to the key ubiquitin modifying enzymes which regulate
membrane and has been shown to deubiquitinate monoubiquitinated mTORC1 a number of specific mTORC2 ubiquitin mediated activators
PDK1 both in vitro and in vivo [193]. However, the role of PDK1 ubi- and repressors have been identified. RICTOR is distinctively ubiquiti-
quitination remains unclear. It is yet to be determined if ubiquitination nated and degraded by SCF-FBXL7 highlighting one of the more inter-
is required for recruitment of PDK1 to the membrane or if PDK1 ubi- esting feedback loops regulating overall PI3K activity. As observed with
quitination is required for AKT activation. a large proportion of ubiquitination reactions catalyzed by E3 ligases,
The mammalian target of rapamycin (mTOR) protein is a serine- ubiquitination is mediated by prior phosphorylation of the substrate.
threonine kinase that belongs to the PI3K family. mTOR is the key Likewise, phosphorylation of RICTOR is prerequisite for its interaction

470
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

to SCF-FBXL7 and eventual degradation. Furthermore, the phosphor- expression and localization of PTEN in a Nedd4-1-deficient mouse
ylation of RICTOR limits RICTOR binding to AKT. The phosphorylation model signifying that NEDD4-1 may not be a relevant E3 ligase for
of RICTOR is mediated by GSK3β which itself is inhibited by AKT PTEN ubiquitination in the murine system [214]. Counteracting the
generating a positive feedback loop when AKT is activated. Meanwhile, effects of NEDD4-1 ubiquitin-specific protease Herpesvirus-associated
GSK3β can also phosphorylate the serine/threonine phosphatase ubiquitin-specific protease (HAUSP), also known as USP7, removes
PHLPP1 promoting the interaction of PHLPP1 with the SCF-βTrCP li- ubiquitin from PTEN in the nucleus resulting PTEN nuclear exclusion
gase complex targeting PHLPP1 for degradation. PHLPP1 depho- [215]. Critically, absence of nuclear PTEN has been linked with tu-
sphorylates AKT limiting the activity of AKT. As such under certain morigenesis [210]. In correlation with PTEN nuclear exclusion, USP7
conditions GSK3β can activate AKT by degrading its negative regulator may therefore be regarded as a potential onco-protein a point further
PHLPP1. Similarly, the deubiquitinating enzyme UCHL1 has been strengthened as USP7 has been observed to be overexpressed in pros-
shown to enhance AKT pathway activation by suppressing the levels of tate cancer [215].
PHLPP1 an effect found to drive the development of lymphoma in vivo X-linked inhibitor of apoptosis protein (XIAP), a RING domain E3
[206]. In contrast, another DUB, USP49, inhibits AKT pathway by ligase has been reported to regulate PTEN stability both in vitro and in
promoting interaction of PHLPP1 to AKT, thus facilitating the de- vivo. XIAP has been shown to enhance PTEN turnover through poly-
phosphorylation of AKT. USP49 does so by deubiquitinating and sta- ubiquitination but similarly ectopic expression of XIAP did not induce
bilizing the scaffold protein FKBP51, which enhances PHLPP1/AKT mono-ubiquitination of PTEN [216]. In contrast, knockdown of XIAP
interaction. using shRNA in 293T and MCF-7 cells reduced both mono, multi-mono
and poly-ubiquitination forms of PTEN resulting in decreased PTEN
5.4. Ubiquitin mediated regulation of PTEN expression in the nucleus followed by a corresponding accumulation of
PTEN in cytoplasm suggesting involvement of another E3 ligase reg-
The lipid phosphatase PTEN plays an indispensable role in con- ulating PTEN mono-ubiquitination. On the other hand, both WWP2
trolling PI3K pathway activation by inhibiting formation of the up- (also known as AIP2), a member of NEDD4 family of E3 ligases and
stream signalling molecule PIP3. PTEN has also recently been shown to CHIP, the chaperone-associated E3 ligase, have been proposed to
negatively regulate EGFR activity by affecting stabilization of the EGFR mediate PTEN degradation and demonstrated to be required for neo-
complex with the ubiquitin ligase CBL [207]. The overall outcome of plastic formation [217,218].
PTEN function in the cytoplasm is the inactivation of downstream on- Along with stability, the phosphatase activity of PTEN has also been
cogenic AKT mediated signalling. Unsurprisingly, post translational demonstrated to be regulated by ubiquitination. Ret Finger Protein
modifications including ubiquitination has been shown to play a huge (RFP) or TRIM27, a member of the tripartite motif (TRIM) family,
role in regulation of PTEN stability and activity as ubiquitinated iso- promotes poly-ubiquitination of PTEN but rather then altering PTEN
forms of PTEN have been reported in a plethora of cancers [208].As stabilization or localization RFP significantly inhibits PTEN phospha-
indicated PTEN is primarily located in the cytosol with only a small tase activity leading to PI3K pathway hyperactivation [219].
fraction of PTEN recruited to plasma membrane to convert PIP3 to Apart from ubiquitination, de-ubiquitination of PTEN has also been
PIP2. This recruitment of PTEN to the membrane is dependent upon on determined to be a critical factor in tumorigenesis. Utilising a genome
an open accessible motif on the surface of PTEN generating a membrane wide library encompassing all the known deubiquitinating enzymes
binding regulatory interface. A phosphorylated C-terminal region of Zhang et al. performed a PTEN interacting DUB screen through which
PTEN forms intra molecular interactions with this interface blocking they identified USP8, USP10, USP13, and USP39 as novel interactors of
PTEN binding to lipid membrane. Upon dephosphorylation of C-term- PTEN [220]. They highlight USP13 as a deubiquitinating enzyme of
inal residues PTEN reverts to the open conformation permitting mem- PTEN whereby USP13 stabilizes PTEN by counteracting ubiquitin-
brane binding and PTEN mediated dephosphorylation of PIP3 to PIP2. mediated degradation of PTEN. Furthermore, they demonstrate that
Importantly, the open conformation of PTEN also leads to nuclear USP13 expression is downregulated in breast cancer correlating with
translocation of the protein where PTEN exerts a tumour suppressive the expectant loss of PTEN expression. OTUD3, plays a similar role in
role in the regulation of DNA repair and genome stability. regulation of PTEN, and tumour progression in breast cancer [221].
Mutations at K13 and K289 of PTEN, are frequently observed in DUBs have has been also shown to control PTEN at transcription level.
Cowdens disease and glioblastoma and are regulatory ubiquitination The DUB Ataxin-3, a Josephin family DUB has been reported to restrict
sites for PTEN function [209,210]. Interestingly, these disease asso- PTEN transcription in lung cancer cells [222].
ciated PTEN mutants display normal phosphatase activity. However, in
both cases the nuclear translocation of the enzyme is perturbed [210]. 6. The role of ubiquitin modifying enzymes in the WNT pathway
The E3 ligase neural precursor cell expressed developmentally down-
regulated protein 4-1 (NEDD4-1) functions in a bi-modal manner to The Wnt signalling pathway plays a key role during embryogenesis/
mediate both mono- and poly-ubiquitination of PTEN with K289 acting development. It has been known to be involved in cell proliferation, cell
as a major site for NEDD4-1 activity. K289 is located within the C2 migration and cell fate determination. Unfortunately, aberrant activa-
domain of PTEN and it has been suggested that mutations at these sites tion of the Wnt pathway is frequently observed in tumorigenesis. The
may disrupt the C2 domain from binding the plasma membrane thus canonical Wnt signalling pathway undergoes activation following Wnt
maintaining high levels of PIP3 [211]. Following the catalyses of PIP3 ligand binding to a seven-pass transmembrane Frizzled (Fz) family of
to PIP2 PTEN is likely polyubiquitinated by NEDD4-1 at K289 to con- receptors and its co-receptors LRP5/6 at the cell surface resulting in the
finement of PTEN in cytoplasm and its subsequent degradation via the recruitment of the adaptor protein Dishevelled to the receptor complex.
proteasome. In contrast, mono-ubiquitination of PTEN by NEDD4-1 at LRP5/6 is subsequently phosphorylated by Casein kinase 1 (CK1) and
K13 and K289 leads to a regulatory modification promoting its nuclear glycogen synthase kinase 3 (GSK3) resulting in a high affinity binding
translocation [212]. Thus, NEDD4-1 acts as both an oncogene and a site for AXIN. This then thwarts AXIN complex mediated phosphor-
tumour-supressor. NEDD4-1 is amplified in a number of cancers and ylation of β-catenin leading to an overall increase in the cytoplasmic
expression of NEDD4-1 correlates with low PTEN levels in lung cancer levels of β-catenin and permitting β-catenin to accumulate in the nu-
[213]. In the majority of cases PTEN associated cancer mutations are cleus where it activates T-cell factor (TCF) and Lymphoid enhancer
present in the C2 domain, however, in some cases the C2 domain re- factor (LEF) leading to the transcription of key target genes such as
mains intact, the latter situation potentially acting as a predictive bio- Cyclin D1 and c-Myc [223,224].
marker for cancers that may respond to NEDD4-1 inhibition. It must be The ubiquitin-proteasome system has previously been described to
noted that data from an independent group revealed no changes in the tightly regulate key components of Wnt pathway (Fig. 6). Cytosolic β-

471
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

Fig. 6. Schematic overview of regulation of Wnt pathway by Ubiquitin ligases and deubiquitinating enzymes. (A) Wnt off: In the absence of Wnt ligand, the Axin-GSK-APC-CK1
destruction complex targets β-catenin for phosphorylation, priming β-catenin for ubiquitination by β-TRCP and resulting in β-catenin degradation. GSK can also target the E3 ligase
RNF220 inhibiting recruitment of USP7 and deubiquitination of β-catenin. USP15 can enhance stabilization of the destruction complex by deubiquitinating APC. The pro Wnt pathway
activator Dishevelled is ubiquitinated by either the E3 ligases ITCH, NEDD4L or CUL-3 reducing its stability and thus limiting Dishevelled recruitment to LRP5/6 and Wnt activation. In
some scenarios Dishevelled can act as a negative regulator of Wnt signalling by binding ZNRF3/RNF43. ZNRF3/RNF43 functions to downregulate Wnt signalling by limiting Frizzled
recruitment to the membrane. ZNRF3/RNF43 also plays a role downstream of β-catenin by sequestering the transcription factor TCF4 to the nuclear membrane. The E3 ligases CBL and
JADE-1 ubiquitinate and destabilise nuclear β-catenin. Taken together multiple ubiquitin modifying enzymes are involved in the downregulation of Wnt signalling.
(B) Wnt on: Upon Wnt ligand binding to the Fzd receptor and the coreceptor Lpr5/6, Dishevelled is recruited to the receptor complex. LRP5/6 is subsequently phosphorylated by CK1 and
GSK resulting in a high affinity binding site for Axin. The binding of Axin to the receptor complex results in the demolition of the destruction complex resulting β-catenin stability and
translocation to the nucleus where it binds to the TCF/LEF family of transcription factors to induce transcription. A number of DUBs act to stabilize the receptor complex including USP8
which deubiquitinates Fzd and USP14 which deubiquitinates Dishevelled. In contrast, the E3 ligases RNF146 and SMURF2 both ubiquitinate and degrade Axin. Axin itself is also regulated
by the SMURF2 homologue SMURF1 however, SMURF1 mediated ubiquitination of Axin results in enhanced K29 ubiquitination of the protein and downregulation of Axin activity.
USP34 also deubiquitinates Axin leading to stabilization of Axin however, the authors also noted that downregulation of USP34 downregulated β-catenin induced transcription of Wnt
target genes. Four DUBs bind and deubiquitinate β-catenin resulting in its stability: USP4, USP9X, USP35 AND USP47. A fifth DUB, USP7, performs a similar function but does so in
complex with RNF220 to limit β-catenin ubiquitination. Also the E3 ligase RAD6B functions in the nucleus to stabilize β-catenin.

catenin and its regulation by Wnt is the essence of Wnt signalling and phosphorylated forms of β-catenin therefore regulating β-catenin sta-
along with its original discovery it was noted that β-catenin levels were bilization in both the Wnt-off and Wnt-on phases [229].
policed by various degradative mechanisms. The AXIN complex is made β-catenin can also be regulated by the E3 ligase MULE with ectopic
up of the scaffolding protein AXIN which contains dispersed binding expression of MULE decreasing β-catenin stabilization. Interestingly,
sites to interact with the tumour suppressor adenomatous polyposis coli MULE appears to function through a Wnt ligand dependent negative
gene product (APC), casein kinase 1 (CK1), and glycogen synthase ki- feedback loop whereby MULE targets both β-catenin and its upstream
nase 3 (GSK3). GSK3 and CK1 coordinate sequential phosphorylation of regulator Dishevelled [230]. As such under low Wnt signalling activa-
β-catenin at S45 (CK1) and subsequently at T41, S37 and S33 (GSK3) tion of MULE is restricted however, under conditions of cellular hy-
[225]. The primary E3 ubiquitin ligase responsible in the regulation of perproliferation promoting constitutive Wnt signalling MULE expres-
β-catenin stability is β-TrCP. GSK3 phosphorylation of β-catenin at S33 sion limits β-catenin stability. In Apcmin mice exhibiting enhanced β-
and S37 generates a binding site for β-TrCP, leading to β-catenin ubi- catenin expression MULE deficiency further accelerated adenoma de-
quitination and degradation [226]. GSK3 and CK1 can also phosphor- velopment compared to Apcmin mutation alone [231].
ylate AXIN and APC increasing the association of these components to Of note, other E3 ubiquitin ligases targeting Dishevelled have also
β-catenin further enhancing β-catenin degradation. been described. The Cullin-3 ubiquitin ligase with the Broad complex,
Another RING finger containing E3 ubiquitin ligase, c-CBL has been tramtrack and Bric- a- Brac (BTB) protein Kelch-like 12 (KLHL12) was
described to ubiquitinate and degrade β-catenin in the nucleus [227]. one of the first E3 ubiquitin ligases found to ubiquitinate and degrade
The authors demonstrate that wild-type c-CBL suppresses while a ligase Dishevelled thereby antagonizing Wnt pathway [232]. Furthermore,
deficient mutant of c-CBL enhances Wnt signalling. Although, the au- two of the HECT domain containing-NEDD4 family of E3 ubiquitin li-
thors describe the UBA domain on c-CBL dimerizes with β-catenin, it gases have been shown to regulate levels of Dishevelled. ITCH ubiqui-
was not until recently that phosphorylation was found to be involved tinates phosphorylated forms of Dishevelled, thereby promoting its
[228]. In contrast, JADE-1 ubiquitylates both phosphorylated and non- proteasome mediated degradation and attenuating Wnt signalling

472
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

[233]. Expectedly, RNAi mediated knockdown of ITCH stabilized Di- ligase RNF146 directly interacts with poly(ADP-ribose) enhancing
shevelled and upregulated Wnt signalling. This group also mapped two tankyrase dependent PARsylation of AXIN promoting its degradation
regions on Dishevelled, a prototypical PPXY motif and DEP domain [241]. AXIN is also regulated by the E3 ligase SMURF2 [242]. SMURF2
through which it interacts with ITCH. Mutation within the PPXY motif ubiquitinates K505 of AXIN leading to protein degradation. AXIN has a
at Y568F or deletion of the DEP domain led to reduced affinity for number of putative PPXY motifs however, it remains undetermined if
ITCH. NEDD4L, which also belongs to the NEDD4 family of HECT the WW domains of SMURF2 are required for this interaction. Similarly,
containing E3 ubiquitin ligases has been shown to target Dishevelled the SMURF2 homologue SMURF1 interaction with AXIN is independent
and promote its proteasome mediated degradation [234]. Interestingly, of its WW domain. Rather the C2 domain of SMURF1 is required for
NEDD4L mediated degradation of Dishevelled involves atypical ubi- K29 linked polyubiquitination of AXIN mediating AXIN SMURF1 loca-
quitin chain formation including K6, K27 and K29 but not the canonical lisation at the plasma membrane, an effect which appears to be cell
K48 ubiquitin chain topology, usually associated with degradation. cycle regulated [243]. Overall, these three E3 ligases RNF146,
Dishevelled can also be regulated through K63 polyubiquitination a SMURF1, and SMURF2 regulate either AXIN stability or function de-
modification regulated by the deubiquitinating enzyme CYLD. CYLD pending on the distinct ubiquitin chain topology.
directly deubiquitinates Dishevelled but the direct function of K63 Opposing the tankyrase-dependent ubiquitination and degradation
ubiquitination or the ligase involved in the process remains unknown. of AXIN is the deubiquitinating enzyme USP34. USP34 was found to
Nevertheless, it remains clear that depletion of CYLD enhanced Wnt associate with AXIN containing protein complexes through liquid
induced β-catenin accumulation in the nucleus and activation of β-ca- chromatography-tandem mass spectrometry. In line with previous re-
tenin induced transcription [235]. Curiously, Dishevelled has also been ports depletion of USP34 led to the polyubiquitnation and degradation
demonstrated to be required for ZNRF3/RNF43 mediated ubiquitina- of AXIN. However, the authors also suggest that loss of USP34 inhibited
tion and degradation of the FZD receptor. Depletion of either Dishev- β-catenin mediated transcription indicating that USP34 may function
elled or ZNRF3/RNF43 enhanced cell surface levels of FZD and LRP5/6 downstream of β-catenin to regulate nuclear accumulation of AXIN
indicating that Dishevelled acts as an intermediary for the recruitment [244]. Although AXIN has been prescribed as a key component of the β-
of these E3 ligase to the receptor complex [236]. This also places Di- catenin destruction complex AXIN is also known to shuttle between
shevelled at a crossroads for both the activation and inhibition of the both the cytoplasm and nucleus where nuclear AXIN expression has
pathway by acting as a scaffold protein required for both AXIN down- been demonstrated to be greatly enriched in diverse cancers. Never-
regulation and FZD stabilization. Further experimentation will no doubt theless, the precise function of how nuclear AXIN enhances β-catenin
be required to tease out the organization of Dishevelled in these two transcription remains unknown. Reflecting on the overall function of
phases of Wnt regulation. ZNRF3/RNF43 can also function downstream the known ubiquitin modifying enzymes that regulate AXIN it becomes
of β-catenin to inhibit Wnt pathway activation by physically interacting apparent that multiple post translational modifications and unique
with the TCF4 transcription factor and tethering TCF4 to the nuclear chain topologies are required to tightly regulate AXIN homeostasis in
membrane [237]. Importantly, oncogenic mutations within RNF43 as- the cells.
sociated with human gastrointestinal tumours disrupted this inhibitory TRABID which belongs to the OTU domain containing family of
mechanism resulting in transactivation of the Wnt pathway. DUBs has been described as a positive regulator of the Wnt signalling
Counteracting these processes, a number of ubiquitin modifying [245]. TRABID binds APC but rather then antagonizing the proteasomal
enzymes have been identified which activate Wnt signalling by reg- turnover of APC TRABID inhibits APC activity through preferential
ulating proteins along multiple nodes in the pathway. USP8/UBPY deubiquitination of K63 linked ubiquitin chains. Furthermore, the effect
along with USP6 were identified as a deubiquitinating enzymes for the of TRABID on APC appears to function downstream β-catenin stabili-
Frizzled receptor. Ubiquitination of receptors at the plasma membrane zation with the authors postulating that APC may function to decrease
induces receptor endocytosis and localization of receptors into lyso- the rate of TCF-β-catenin complex formation, a process seemingly de-
somes. Within these lysosomes receptors can either be recycled back to pendent upon K63 ubiquitination of APC and one which is reverted by
cell membrane or targeted for degradation in a ubiquitin dependent TRABID.
manner. USP6 and USP8 were found to deubiquitinate FZD stimulating Recently, Ubiquitin specific protease 4 (USP4) was identified as a
FZD recycling and increase FXD expression at the plasma membrane DUB targeting β-catenin. Deubiquitination of β-catenin by USP4 re-
[238,239]. Another deubiquitinating enzyme that has been shown to verses the ubiquitination mediated degradation of β-catenin, upregu-
positively regulate Wnt signalling is USP14. Interestingly, both USP14 lating Wnt signalling. Unsurprisingly, a positive co-relation between
and the aforementioned CYLD deubiquitinate K63 polyubiquitinated levels of USP4 and β-catenin was observed in tissues from patients di-
isoforms of Dishevelled. However, in contrast to CYLD, which acts as a agnosed with colorectal cancer [246]. In a separate study, USP4 was
negative regulator of the pathway, USP14 positively enhanced Di- also identified in an RNAi screen performed in the colorectal cancer cell
shevelled function as genetic and chemical suppression of USP14 im- line SW480 using the Wnt-reporter TOPFlash reporter assay [247].
paired downstream Wnt signalling. This disparity maybe explained by However, in contrast to the previous work RNAi depletion of USP4
the targeting of specific ubiquitination sites by both enzymes. CYLD caused an up-regulation of Wnt signalling. In this case, USP4 was ob-
deubiquitinates K46 and K50 within the DIX domain of Dishevelled served to interact with Wnt regulators Nemo-like kinase (NLK) and
which mediates the dynamic polymerization of the protein enhancing TCF4. USP4 deubiquitinated a subpopulation of TCF4. However, it re-
binding sites for Wnt signalling partners [240]. This suggests that K63 mains unclear if TCF4 ubiquitination is required for TCF4/LEF medi-
mediated ubiquitination at K46 and K50 is required for polymerization ated transcription. Interestingly, NLK promoted nuclear accumulation
to occur for the binding to Wnt signalling partners, potentially AXIN, an USP4. As NLK positively regulates Wnt signalling it appears that either
effect downregulated by CYLD. In contrast, USP14 deubiquitinates NLK regulates USP4 nuclear localization as part of negative feedback
K444 and K451 within the DEP domain of Dishevelled, a motif required loop to downregulate TCF or that NLK enhances USP4 mediated sta-
membrane translocation for the protein. Thus, K63 ubiquitination at bilization of β-catenin further promoting TCF mediated transcription.
these residues in the DEP domain appears to have a negative regulatory Even though, both the studies were performed using colorectal cancer
role by inhibiting the recruitment of Dishevelled to the membrane. cell lines, caution must be taken to carefully understand the role of
A number of components of the AXIN-APC-GSK-CK1 destruction USP4 in both scenarios.
complex are also regulated by ubiquitination. AXIN stability is pri- USP15 and USP47 have also been identified as positive regulators of
marily regulated by the poly-ADP-ribosylating enzymes tankyrase 1 and Wnt signalling. Both USP15 and USP47 form a complex with β-catenin
tankyrase 2, whereby both enzymes interact with a highly conserved leading to its deubiquitination and stabilization [248,249]. USP7, a
domain of AXIN targeting AXIN for degradation. In this setting the E3 closely related member to USP47 has been shown to function along

473
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

with an E3 ubiquitin ligase RNF220 to stabilize β-catenin. Although degradation of oxidized proteins and enhanced resistance towards
RNF220 is an E3 ubiquitin ligase, knockdown of USP7 renders the oxidative stress [254].
former inactive. Knockdown of either RNF220 or USP7 impairs overall UCH-L1 has been linked to Parkinson's disease and has been iden-
Wnt signalling in colon cancer cells [250]. tified as a biomarker for aggressive multiple myeloma and enhances
Like the β-catenin oppressor MULE, RAD6B is a transcriptional HIF-1 mediated metastasis [257,258]. LDN-57444, a molecule which
target of β-catenin and functions through a feedback loop to regulate belongs to a class of isatin O-acyl oximes was identified by a high
overall β-catenin levels. However, unlike MULE, RAD6B mediated throughput screen to find inhibitors against UCH-L1. Though the mo-
polyubiquitnation of β-catenin renders β-catenin insensitive to protea- lecule can inhibit UCH-L1, it was found that proliferation was increased
somal degradation. RAD6B is a 17 kDA ubiquitin conjugating enzyme in H1299 lung tumour cell line though this effect was not exhibited a in
which induces K63 linked β-catenin polyubiquitination. Depletion of lung tumour lines that did not express UCH-L1. RNAi based approaches
RAD6B decreases β-catenin polyubiquitnation and stability limiting β- also had similar proliferative effects and suggests that UCH-L1 enzy-
catenin transcription. Unsurprisingly, β-catenin and RAD6B expression matic activity is antiproliferative and UCH-L1 expression may be a
is positively correlated in breast carcinoma with RAD6B levels sig- adaptive response to enhanced tumour growth [259]. In a second study,
nificantly enhanced in breast cancer compared to normal controls. LDN-57444 treatment enhanced apoptosis by increasing the levels of
Taken together it becomes wholly apparent that a multitude of post highly ubiquitinated proteins activating unfolded protein response, a
translational modifications are required for effective regulation of Wnt protective response that promotes apoptosis when cellular stress con-
signalling with manipulation of ubiquitin modifying enzymes offering tinues to persist [260]. Similarly, LDN91946, a moderately potent in-
an exciting opportunity for cancer therapy and regenerative medicine. hibitor of UCH-L1 was identified using an assay measuring the hydro-
lysis of the fluorogenic substrate Ub-AMC. Subsequent work analyzing
7. Therapeutic intervention of dubs structural activity relationship studies and kinetics revealed that this
compound is a noncompetitive inhibitor of UCH-L1 but not other cy-
Following the approval of Bortezomib, a proteasome inhibitor, as an steine hydrolases tested including such as UCH-L3, USP5 and caspase 3
anti-cancer therapeutic for the treatment of multiple myeloma, in- [261]. Again, downregulation of UCH-L1 by LDN91946 enhanced cell
hibitors of the ubiquitin-proteasome system (UPS) has received much death indicating that under certain conditions downregulation of UCH-
attention and focus as a potential and attractive drug target for cancer L1 may be used to optimize patient responses [262].
therapy. However, to date no inhibitors targeting the ubiquitin pro- USP7 is probably the most well studied of all the known deubiqui-
teasome system other than those that inhibit proteasome function have tinating enzymes targeting a plethora of cancer relevant genes in-
been approved for clinical use. Deubiquitinating enzymes of course are cluding both tumour suppressors (p53, PTEN) and oncogenes (MDM2,
an integral part of the UPS, functioning to remove ubiquitin moieties IRS1). Nevertheless, USP7 may be considered a promising therapeutic
from polyubiquitin chains or target proteins. The intrinsic isopeptidase target in cancer. With the help of biochemical assays and activity based
activity of DUBs results in the cleavage of the carboxyl terminus of protein profiling small molecule antagonists with high specificity have
ubiquitin destabilizing ubiquitin substrate binding. Taking into account been identified against USP7. Both P02207 and P5091 target USP7 with
that the majority of cancers demonstrate enhanced expression of DUBs the added function that P5091 simultaneously targets USP47 directing
which ostensibly act as oncogenes in a number of cancer relevant the cell fate towards apoptosis. Interestingly P5091 exhibited this effect
pathways, DUB inhibition may be a considered as a valid cancer ther- in multiple myeloma cells resistant to bortezomib suggesting that
apeutic strategy. The majority of the DUBs are cysteine proteases (the P5091 may be used in bortezomib resistant myeloma patients as pre-
exception being the JAMM family), wherein the catalytic cysteine re- liminary results indicate that P5091 is well tolerated in animal models
sidues sheltered in the active sites of DUBs are highly reactive towards and inhibits tumour growth and prolongs survival [263,264]. In-
electrophiles. Most ubiquitination reactions involve the formation of cidentally, blocking HDM2 and p21 abrogated the cytotoxicity induced
thioester bonds between the c-terminal tail of ubiquitin and its sub- by P5091. Further research has led to the development of a potent
strate whereby the nucleophile cysteine containing thiol side chains can analogue of P5091 called cdp14, against both USP7 and USP47, which
then attack this reaction. Revolving around this property, most of the has enhanced potency, solubility and a strong metabolic reactivity
DUB inhibitors that contain α, β-unsaturated ketones have been de- profile [264]. Finally, HBX19818 has also been identified as a USP7
monstrated to possess DUB inhibitory activity by trapping these thiol inhibitor. Interestingly, HBX19818 covalently modifies the active
side chains. Towards the development of such DUB inhibitors, a wide Cys233 residue of USP7 limiting overall activity of the enzyme [265].
range of compounds ranging from synthetic small molecules to natural Colon cancer cells treated with this molecule produced a G1 arrest
products with DUB inhibitory activity have been isolated. This section mimicking results observed in cell lines following USP7 shRNA
reviews effective strategies to target DUBs. We would also like the knockdown, showcasing its high anti tumorigenic potential [265].
readers to refer to the comprehensive review by D'Arcy et al. [251]. The USP1/UAF complex is involved in the translesion synthesis
through the deubiquitination of FANCD2 and FANCI [266]. Two im-
7.1. Small molecule DUB inhibitors portant domains in UAF1 regulate FANC pathway activation. The SLD2
region of UAF1 binds FANCI and the WD40 domain of UAF1 binds and
As highlighted in this review, DUBs catalyze the removal of ubi- stimulates USP1. Disruption of either of these domains promotes
quitin moieties from substrates. For the most part the eviction of such FAND2/FANCI ubiquitination and enhanced DNA repair defects [267].
ubiquitin moieties results in enhanced protein stability. However, a Utilising a high throughput screen, pimozide, an anti-psychotic drug
number of DUBs (USP14, RPN11, UCH-L5) are also physically and and GW7647, a PPAR-α-agonist were shown to act as potent inhibitors
functionally associated with the 26S proteasome and act in a timely and of USP1/UAF activity [268]. These inhibitors have been shown to
substrate specific manner to remove ubiquitin for proper substrate target the USP and WD40 repeat protein complex. Furthermore, pi-
processing, and as such targeted inhibition of proteasome associated mozide and GW7647 act synergistically with cisplatin in inhibiting
DUBs may mimic the effect of bortezomib [252,253]. A small molecule proliferation of non-small cell lung cancer cells resistant to cisplatin
inhibitor of the DUB USP14 was identified through a high through put [268].
screen called IU1 (inhibitor of USP14) [254]. This molecule, when LS1 was also recently identified as a potent inhibitor of UCH-L3.
exposed to cells, enhanced degradation of several proteasome sub- Applying a FRET based approach the authors analyzed the synthesis of
strates implicated in neurodegenerative disease, as USP14 is essential ubiquitin bioconjugates to substrates [269]. Ubiquitinated peptides
for the maintenance of synaptic ubiquitin levels and neuromuscular were labelled with a pair of FRET labels and were used to screen a
junction development [255,256]. USP14 inhibition led to quicker library comprising about 1000 compounds against UCH-L3.

474
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

One of the first DUB inhibitors PR-619, is a broad spectrum DUB inhibitors compared to the other members of the A, B or E pros-
inhibitor which leads to an overload of ubiquitinated proteins, protein taglandin series. PGJ2, Δ12-PGJ2 and 15Δ-PGJ2 are sequentially me-
aggregate formation and subsequent inhibition of the UPS. PR-619 also tabolized products of PGD2 [278–280]. PGs such as Δ12-PGJ2 and Δ7-
leads to the activation of the autophagy pathway by inducing the PGA1 methyl ester have antitumour and antiviral activities and are
transport of lysosomes to the aggregates formed [270,271]. However, easily taken up by cells and accumulate in the nuclei via covalent in-
due to its broad spectrum activity PR-619 is highly toxic in vivo which teractions that ultimately lead to growth inhibition. α, β- unsaturated
has therefore limited its effect in clinical development. ketones are highly susceptible to nucleophilic compounds with thiols as
they are particularly strong nucleophiles that PGs react with en-
7.2. Michael acceptors zymatically or non-enzymatically. Specifically, these cyclopentone PGs
interact with the thiol nucleophiles at the β position within the cyclo-
The formation of covalent bonds is an important criterion for the pentenone ring. Aside from acting as Michael acceptors another pos-
design of anticancer molecules. Most antineoplastic agents bind directly sible mechanism through which these prostaglandins can lead to UPS
to their substrates. However, most these agents lack target selectivity. inhibition is through protein carbonylation and oxidative stress induced
Michael acceptors, on the other hand can be structurally modified to by cyclopentone PGs which eventually leads to the decrease in activities
react selectively with specific nucleophiles. The Michael reaction of S6 ATPase and 26 proteasome [281]. This oxidative stress arises
comprises both Michael acceptors and Michael donors, and comprises when reactive oxygen species (ROS) are generated and can have po-
of a nucleophilic addition of a carbanion or other types of nucleophiles. tential damaging effects on cellular components. Interestingly, UCHL1
The α, β- unsaturated compounds (ketone) undergoing Michael addi- is one such protein which is oxidized in cells treated with 15Δ-PGJ2.
tion is called the Michael acceptor and the highly reactive nucleophiles Δ12-PGJ2 and 15Δ-PGJ2 are potent cyclopentone PGs and have been
(cysteines) are referred to as Michael donors and together their product reported to inhibit UCH-L3 and UCH-L1 respectively [282,283]. Ad-
is referred to as the Michael adduct. The 1,4 conjugate reaction of α, β- ditionally, cyclopentone PGs induced unfolding and aggregation of
unsaturated carbonyl compounds and nucleophiles is referred to as the UCH-L1, thus inhibiting its enzyme activity [284]. UCH-L1 is abundant
Michael reaction. In case of DUB inhibitors, the addition of the Michael in the brain with its expression particularly localized to the Lewy bodies
acceptor traps the catalytic cysteine of the DUB active site rendering the or the neurofibrillary tangles. Furthermore, UCH-L1 has been docu-
targeted DUBs inactive. mented to be involved in a ubiquitin-dependent proteolytic pathway
WP1130 (Degrasyn) is a small molecule, selective deubiquitinase and has been implicated in the pathogenesis of Parkinson's disease. A
inhibitor of both USP and UCH subclasses. WP1130 is derived from a single thiol group on Cys152 of UCH-L1, irrespective of the presence of
compound with a Janus-activated kinase 2 (JAK2) inhibitor activity. 5 other cysteine groups in UCH-L1, reacts with the α, β- unsaturated
WP1130 inhibits DUBs such as USP5, USP9X, USP14, UCH-L1 and carbonyl center in the cyclopentenone rings of PG leading to the for-
UCH37, all of which are known to regulate survival protein stability. mation of a covalent adduct destroying its native structure and even-
WP1130 also suppresses BCR/ABL, is a JAK2 transducer and activator tually its DUB activity [284]. Notably, 15Δ-PGJ2 also inactivates p53
of transcription factor STAT. The addition of WP1130 leads to rapid function by its binding to Cys277 reducing significantly its binding and
accumulation of polyubiquitinated K48/K63-linked proteins into jux- transcriptional activity. As discussed the majority of the DUBs are cy-
tanuclear aggresomes, independent of 20S proteasome activity. steine proteases which are highly reactive nucleophiles and cyclo-
Furthermore, WP1130 inhibits tumour activated DUBs and results in pentone PGs have Micheal acceptors attributing to the inhibitory effect
the downregulation of anti-apoptotic proteins and upregulation pro- of this set of compounds towards DUBs.
apoptotic proteins such as MCL-1 and p53 [272–274]. Preliminary data
has also indicated that it may be clinically relevant as WP1130 in 7.4. Chalcone inhibitors
combination with bortezomib had anti-tumour activity in mantle cell
lymphoma animal models [275]. Chalcone or chalconids is an aromatic ketone and an enone that
Eeyarestatin-1(Eer1) is an inhibitor of the endoplasmic reticulum forms the central core for a plethora of critical biological compounds.
associated protein degradation (ERAD) pathway [276]. Proteins that Chalcone compounds such as G5, b-AP15 and RA-9 have been described
are terminally misfolded, are recognized by chaperones on the en- to inhibit intrinsic deubiquitinase activity. Chalcones contain cross
doplasmic reticulum and are transported to depots for ubiquitination conjugated α,β-unsaturated ketones and accessible β-carbons and in-
and proteasomal degradation through the ERAD pathway. Eeyarestatin- hibit DUB activity but are unrelated to PGs. Chalcone DUB inhibitors
1 binds directly to p97 ATPase and acts by blocking the degradation of can be highly specific such as b-AP15 or highly broad spectrum like G5.
misfolded proteins associated with the p97-associated deubiquitinating b-AP15 and its analogue VLX1570 are other proteasome associated
complex in cells preventing ataxin-3 mediated deubiquitination of DUB inhibitors involved in the specific inhibition of USP14 and UCH-
substrates [277]. Notably, eeyarestatin-1 has exhibited anti-cancer L5. b-AP15 was identified in cell based screens for compounds enhan-
properties similar to that of bortezomib. cing cellular apoptosis independent of either cathepsin D or p53 func-
tion [285,286]. Dual inhibition of UCH-L5 and USP14 using RNA in-
7.3. Cyclopentone prostaglandins terference not only led to the accumulation of proteasomal substrates
but led to loss of cell viability [287,288]. Furthermore, b-AP15 has been
Cyclopentone prostaglandins are a subset of prostaglandins (PGs) or reported to show anti-neoplastic properties in animal models, multiple
eicosanoids that induce the accumulation of polyubiquitinated proteins myeloma and solid tumours [287,289]. Interestingly, using gene ex-
in cells and correspondingly inhibit DUB activity [278]. These eicosa- pression and connectivity MAP analysis showed that b-AP15 induced a
noids are characterized by their electrophilic nature and their dis- similar gene expression profile to that of the J series prostaglandin 15Δ-
tinctive cross conjugated α, β-unsaturated ketone which act as Michael PGJ2 [289].
acceptors. Both A series and J series prostaglandins are capable of in- Two small molecules namely G5 and F6 were identified in a small
activating wildtype p53 by impairing the conformation and phosphor- chemical library screen that was used to identify alternative pathways
ylation of p53 resulting in the downregulation of p53 transcriptional of caspase activation. The two molecules were capable of activating an
activity [278]. However, the J series of prostaglandins contain a exo- apoptosome-independent apoptotic pathway by targeting the UPS and
cyclic α, β-unsaturated ketone, a unique structural determinant that inhibiting ubiquitin isopeptidase activity [290,291]. Both inhibitors
confers a pro-apoptotic effect through the inhibition of ubiquitin iso- upregulate the BH3-only protein NOXA, and the inhibitor of apoptosis
peptidase activity regardless of p53 mediated transcriptional suppres- antagonist SMAC [290]. NOXA has been shown to play acritical role in
sion [278]. Additionally, prostaglandins of the J series are more potent the induction of mitochondrial fragmentation and caspase activation

475
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

[291]. F6 in particular was reported by another group to be associated inhibitory activity. Curcumin is a diarylheptanoid, belonging to the
with the inhibition of USP2 and USP7 and the inhibition of the SENP2 family of cucuminoids, a natural phenol, bestowing the yellow colour of
deSUMOylase [292]. turmeric. Furthermore, curcumin is a homodimer of feruloylmethane,
Some other known partially selective DUB inhibitors, which happen containing a methoxy and a hydroxyl pharmacophore. Additionally,
to fall in the chalcone family are AM146, RA-9, RA-14 and RAMB1 curcumin is an extensively studied phytochemical with anti-cancer,
[293,294]. These DUB inhibitors induce the rapid accumulation of anti-microbial, anti-inflammatory and anti-oxidant properties to name
polyubiquitinated substrates depleting the pool of free ubiquitin a few. Studies also show that the anti-tumour property of curcumin
without affecting the 20S proteasome. The functionality of these novel arises from the presence of two α,β-unsaturated ketone moieties.
small molecule inhibitors is based on the presence of a α,β-unsaturated Curcumin was observed to induce the accumulation of poly-
carbonyl group in the inhibitor making it susceptible to the nucleophilic ubiquitinated proteins at high concentrations, through a shotgun pro-
attack by the sulfhydryl moieties on the cysteines present in the active teomic approach [300]. This group also showed that curcumin down-
sites of DUBs. These chalcone DUB inhibitors directly suppresses the regulated some of the proteins associated with cellular assembly,
DUB activity of UCH-L1, UCH-L3, USP2, USP5 and USP8 which are organization, biosynthesis and glycolysis. Notably, the accumulation of
particularly known to play key roles in the turnover and the stability of polyubiquitinated oncogenes such as NF-κB and cyclin D1 potentially
critical regulators of cell survival and proliferation. Additionally, these indicates the mechanism of action of this intriguing compound.
compounds down regulate cyclin D while concomitantly upregulating AC17, a 4-arylidene curcumin analog, has been reported to inhibit
p53, p27 and p16 leading to arrest in S-G2/M phase of the cell cycle. the deubiquitinase activity of 19S ribosomal particles. Though AC17 is
Furthermore, these inhibitors, abrogate anchorage independent growth a synthesized product, it acts as an inhibitory kB kinase inhibitor (IκK)
and promote the onset of apoptosis in ovarian, breast and cervical and leads to a rapid accumulation of ubiquitinated proteins without
cancer cell lines, displaying no significant alterations in primary human inhibiting the proteasome proteolytic activities. It acts differently from
cells [293,294]. its parent compound curcumin which is a proteasome proteolytic in-
hibitor by serving as an irreversible deubiquitinase inhibitor of 19S
7.5. Natural compounds exhibiting DUB inhibitory activity ribosomal particles, leading to the inhibition of the NF-κB pathway and
reactivation of the proapoptotic protein p53. Treatment with AC17 has
Many compounds affect the ubiquitin proteasome system without been demonstrated to suppress tumour growth in lung xenograft model
the direct inhibition of the proteasome activity, chiefly brought about [301].
by compounds falling under the umbrella of DUB inhibitors.
Interestingly, a number of natural products exist exhibiting DUB in- 7.6. Ubiquitin variant inhibitors
hibitory activities.
Gambogic acid, a xanthanoid with cytotoxic properties, is isolated As highlighted in this review, ubiquitination of substrates by E3
from the resin of Garcinia hanburyi. Gambogic acid has an α,β-un- ligases occurs through the formation of an isopeptide bond between the
saturated ketone moiety which directs growth inhibition and the in- C-terminal carboxyl group of ubiquitin and the lysine of the substrate, a
duction of apoptosis in cancer cells. Gambogic acid has been used in process which is reversed through the catalytic properties of DUBs.
traditional Chinese medicine for ages and has been shown to have cy- Despite their low sequence similarity, USP catalytic domains share a
totoxic activity on several cancer cells [295]. Gambogic acid displays common fold that includes a large structurally conserved ubiquitin
cytotoxic activity by inducing changes in gene expression profiles and binding site for the targeted ubiquitin.
affects the ubiquitin proteasome system. Gambogic acid also lead to Ubiquitin binds with low affinity to these ubiquitin binding domains
enhanced polyubiquitination potentially through the inhibition of (UBDs) a evolutionary consequence which permits ubiquitin to bind to
chymotrypsin activity associated with the 20s proteasome leading to all UBDs equally. Since ubiquitin binds in general with low affinity to
the induction of apoptosis and cell death. Interestingly, the gene sig- USPs but through a large contact area specific alterations within the
nature profiles of cells exposed to gambogic acid show similar profiles ubiquitin molecule itself would result in potential increased binding
as cells exposed to UPS inhibitors like PGJ2, celastrol and with aferinA affinity of the altered ubiquitin to the ubiquitin binding domain of the
marking its potential to inhibit DUB activity [296]. USPs. Furthermore, as each USP displays a relatively sequence specific
Betulinic acid is a naturally occurring pentacyclic triterpenoid, that UBD any ubiquitin variant should be USP specific.
has been isolated from many diverse plants like white birch (Betula Tight binding of these ubiquitin variants should therefore act as
pubescens), ber tree (Zizyphus mauritina), carpenters herb (Prunella vul- competitive inhibitors of catalytic activity by blocking the recognition
garis), rosemary and tropical carnivorous plants such as Triphyophyllu of endogenous ubiquitinated substrates. Using this as a backdrop, Sidhu
peltatum and Ancistrocladus heyneanus. Betulinic acid has a variety of and colleagues generated combinatorial, phage-displayed libraries of
biological and medicinal properties including antiviral, antibacterial, ubiquitin variants and effectively identified inhibitors targeting USP8,
anti-inflammatory, antihelmintic and antioxidant properties. USP21, USP2a, USP7, USP10 and OTUB1 [302,303]. In all of the cases
Additionally, betulinic acid has been reported to show significant cy- each of the ubiquitin variants bound to their respective ubiquitin
totoxic effect on several cell lines and even human melanoma xenograft binding domains with a greater efficiency then wild type ubiquitin and
animal models [297]. A 20% betulinic acid ointment has been used in more importantly did not cross react with any of the other USPs tested.
the treatment of atypical moles (Dysplastic nevi) [298]. Interestingly, Importantly, ubiquitin variants targeting OTUB1 selectively inhibited
the anticancer property of betulinic acid is carried out by inducing OTUB1 mediated cleavage of K48 di-ubiquitin. Although, ubiquitin
apoptotic cell death in cancer cells by triggering apoptosis through the variant inhibitors cannot be used in patients due to their relative size
inhibition of transmembrane potential in isolated mitochondria [299]. 8 kDa ubiquitin variant technology does allow potent inhibition of
Betulinic acid inhibits multiple DUBs, resulting in the overall accumu- targeted USPs and can be utilized for future target validation and drug
lation of polyubiquitinated proteins and resulting in the decreased le- discovery for all UBD proteins.
vels of oncoptoteins [299]. A similar effect was also observed in TRAMP
transgenic mice model of prostate cancer, wherein betulinic acid in- 8. Concluding remarks
hibited primary tumours by increasing apoptosis and decreasing an-
giogenesis through a marked decrease in androgen receptor and cyclin Since its identification over 35 years ago our understanding of
D1 protein levels [299]. ubiquitin and the enzymes that manipulate its behavior has improved
Curcumin, a principal component of turmeric (Curcuma longa), a dramatically. We now know that ubiquitination plays a central role in
prominent spice ingredient used in most south Asian dishes has DUB virtually all biological processes including a number pathways involved

476
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

in neoplastic disease. Following on from the overall success of borte- [11] H.M. Oosterkamp, E.M. Hijmans, T.R. Brummelkamp, S. Canisius, L.F. Wessels,
W. Zwart, R. Bernards, USP9X downregulation renders breast cancer cells resistant
zomib expectation was high to develop other specific drugs targeting to tamoxifen, Cancer Res. 74 (2014) 3810–3820.
components of the ubiquitin system. However, for the most part these [12] H. Potu, L.F. Peterson, M. Kandarpa, A. Pal, H. Sun, A. Durham, P.W. Harms,
expectations have not been met. In the majority of cases these dis- P.C. Hollenhorst, U. Eskiocak, M. Talpaz, N.J. Donato, Usp9x regulates Ets-1
ubiquitination and stability to control NRAS expression and tumorigenicity in
appointing results are not necessarily attributed to the fact that these melanoma, Nat. Commun. 8 (2017) 14449.
inhibitors are not specific towards their respective targets but rather [13] M. Schwickart, X. Huang, J.R. Lill, J. Liu, R. Ferrando, D.M. French, H. Maecker,
because both E3s and DUBs target multiple protein complexes. K. O'Rourke, F. Bazan, J. Eastham-Anderson, P. Yue, D. Dornan, D.C. Huang,
V.M. Dixit, Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell
Therefore, a better understanding of ubiquitin mediated signalling is survival, Nature 463 (2010) 103–107.
critical to designing effective therapeutic strategies. A number of key [14] W. Ouyang, S. Zhang, B. Yang, C. Yang, J. Zhang, F. Zhou, C. Xie, Beta-catenin is
questions still remain which if answered may ultimately lead to the regulated by USP9x and mediates resistance to TRAIL-induced apoptosis in breast
cancer, Oncol. Rep. 35 (2016) 717–724.
identification novel therapeutic targets. For example, what are the
[15] H. Thanh Nguyen, D. Andrejeva, R. Gupta, C. Choudhary, X. Hong, P.J. Eichhorn,
mechanisms behind the specific targeting of E3 ligases and DUBs to A.C. Loya, S.M. Cohen, Deubiquitylating enzyme USP9x regulates hippo pathway
substrates? In the majority of cases phosphorylation precludes ligase activity by controlling angiomotin protein turnover, Cell Discov. 2 (2016) 16001.
recruitment but this is observed in only a proportion of cases. [16] P.A. Perez-Mancera, A.G. Rust, L. van der Weyden, G. Kristiansen, A. Li,
A.L. Sarver, K.A. Silverstein, R. Grutzmann, D. Aust, P. Rummele, T. Knosel,
Furthermore, what is the effect of post-translational modifications on C. Herd, D.L. Stemple, R. Kettleborough, J.A. Brosnan, A. Li, R. Morgan, S. Knight,
the activity of ubiquitin modifying enzymes themselves and finally, J. Yu, S. Stegeman, L.S. Collier, J.J. ten Hoeve, J. de Ridder, A.P. Klein,
what is the role of non-canonical ubiquitin chains in substrate signal- M. Goggins, R.H. Hruban, D.K. Chang, A.V. Biankin, S.M. Grimmond, I. Australian
Pancreatic Cancer Genome, L.F. Wessels, S.A. Wood, C.A. Iacobuzio-Donahue,
ling? New and emerging technologies such as high resolution crystal- C. Pilarsky, D.A. Largaespada, D.J. Adams, D.A. Tuveson, The deubiquitinase
lography, CRISPR/CAS9 screens, and affordable Next-Gen sequencing USP9X suppresses pancreatic ductal adenocarcinoma, Nature 486 (2012)
along with highly specific ubiquitin modifying enzyme inhibitors will 266–270.
[17] E.W. Harhaj, V.M. Dixit, Deubiquitinases in the regulation of NF-kappaB signaling,
undoubtedly help tease out the roles of these enzymes in cancer pro- Cell Res. 21 (2011) 22–39.
gression and other diseases and ultimately pave the way for better [18] P.M. Siegel, J. Massagué, Cytostatic and apoptotic actions of TGF-β in homeostasis
therapies. and cancer, Nat. Rev. Cancer 3 (2003) 807–820.
[19] J. Massagué, TGFβ in cancer, Cell 134 (2008) 215–230.
[20] J. Seoane, Escaping from the TGFβ anti-proliferative control, Carcinogenesis 27
Acknowledgments (2006) 2148–2156.
[21] Y. Shi, J. Massagué, Mechanisms of TGF-β signaling from cell membrane to the
nucleus, Cell 113 (2003) 685–700.
The authors thank all the members in the Eichhorn Lab at CSI
[22] J. Groppe, C.S. Hinck, P. Samavarchi-Tehrani, C. Zubieta, J.P. Schuermann,
Singapore for scientific discussions on the topics presented here. The A.B. Taylor, P.M. Schwarz, J.L. Wrana, A.P. Hinck, Cooperative assembly of TGF-β
authors apologize to the authors whose papers we could not cite due to superfamily signaling complexes is mediated by two disparate mechanisms and
space limitations. distinct modes of receptor binding, Mol. Cell 29 (2008) 157–168.
[23] J. Massagué, TGF-β signal transduction, Annu. Rev. Biochem. 67 (1998) 753–791.
[24] J. Massague, J. Seoane, D. Wotton, Smad transcription factors, Genes Dev. 19
Disclosure statement (2005) 2783–2810.
[25] C. Alarcon, A.I. Zaromytidou, Q. Xi, S. Gao, J. Yu, S. Fujisawa, A. Barlas,
A.N. Miller, K. Manova-Todorova, M.J. Macias, G. Sapkota, D. Pan, J. Massague,
No potential conflict of interest was reported by the authors. Nuclear CDKs drive Smad transcriptional activation and turnover in BMP and TGF-
beta pathways, Cell 139 (2009) 757–769.
Funding [26] R.S. Lo, J. Massague, Ubiquitin-dependent degradation of TGF-[bgr]-activated
Smad2, Nat. Cell Biol. 1 (1999) 472–478.
[27] H. Zhu, P. Kavsak, S. Abdollah, J.L. Wrana, G.H. Thomsen, A SMAD ubiquitin
This work was in part supported by the by the National Research ligase targets the BMP pathway and affects embryonic pattern formation, Nature
Foundation Singapore and the Singapore Ministry of Education under 400 (1999) 687–693.
[28] R.S. Lo, J. Massague, Ubiquitin-dependent degradation of TGF-beta-activated
its Research Centers of Excellence initiative, Koninklijke Philips N·V smad2, Nat. Cell Biol. 1 (1999) 472–478.
and the Ministry of Education Academic Research Fund Tier 1 grants [29] X. Lin, M. Liang, X.-H. Feng, Smurf2 is a ubiquitin E3 ligase mediating proteasome-
(T1-2013 Sep-10) and (T1-2014 Oct-08). dependent degradation of Smad2 in transforming growth factor-β signaling, J.
Biol. Chem. 275 (2000) 36818–36822.
[30] P. Kavsak, R.K. Rasmussen, C.G. Causing, S. Bonni, H. Zhu, G.H. Thomsen,
References J.L. Wrana, Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the
TGFβ receptor for degradation, Mol. Cell 6 (2000) 1365–1375.
[1] R.C. Kane, A.T. Farrell, R. Sridhara, R. Pazdur, United States Food and Drug [31] S. Bonni, H.-R. Wang, C.G. Causing, P. Kavsak, S.L. Stroschein, K. Luo, J.L. Wrana,
Administration approval summary: bortezomib for the treatment of progressive TGF-β induces assembly of a Smad2–Smurf2 ubiquitin ligase complex that targets
multiple myeloma after one prior therapy, Clinical Cancer Research: An Official SnoN for degradation, Nat. Cell Biol. 3 (2001) 587–595.
Journal of the American Association for Cancer Research 12 (2006) 2955–2960. [32] Y. Zhang, C. Chang, D.J. Gehling, A. Hemmati-Brivanlou, R. Derynck, Regulation
[2] F. Bernassola, M. Karin, A. Ciechanover, G. Melino, The HECT family of E3 ubi- of Smad degradation and activity by Smurf2, an E3 ubiquitin ligase, Proc. Natl.
quitin ligases: multiple players in cancer development, Cancer Cell 14 (2008) Acad. Sci. 98 (2001) 974–979.
10–21. [34] X. Lin, M. Liang, X.H. Feng, Smurf2 is a ubiquitin E3 ligase mediating proteasome-
[3] R.J. Deshaies, C.A. Joazeiro, RING domain E3 ubiquitin ligases, Annu. Rev. dependent degradation of Smad2 in transforming growth factor-beta signaling, J.
Biochem. 78 (2009) 399–434. Biol. Chem. 275 (2000) 36818–36822.
[4] M.B. Metzger, V.A. Hristova, A.M. Weissman, HECT and RING finger families of E3 [35] H.I. Chen, M. Sudol, The WW domain of Yes-associated protein binds a proline-
ubiquitin ligases at a glance, J. Cell Sci. 125 (2012) 531–537. rich ligand that differs from the consensus established for Src homology 3-binding
[5] V. Kirkin, I. Dikic, Ubiquitin networks in cancer, Curr. Opin. Genet. Dev. 21 (2011) modules, Proc. Natl. Acad. Sci. U. S. A. 92 (1995) 7819–7823.
21–28. [36] G. Murakami, T. Watabe, K. Takaoka, K. Miyazono, T. Imamura, Cooperative in-
[6] Y. Ye, M. Rape, Building ubiquitin chains: E2 enzymes at work, nature reviews, hibition of bone morphogenetic protein signaling by Smurf1 and inhibitory Smads,
Mol. Cell. Biol. 10 (2009) 755–764. Mol. Biol. Cell 14 (2003) 2809–2817.
[7] R. Yau, M. Rape, The increasing complexity of the ubiquitin code, Nat. Cell Biol. [37] L.Y. Tang, M. Yamashita, N.P. Coussens, Y. Tang, X. Wang, C. Li, C.X. Deng,
18 (2016) 579–586. S.Y. Cheng, Y.E. Zhang, Ablation of Smurf2 reveals an inhibition in TGF-beta
[8] Y. Kulathu, D. Komander, Atypical ubiquitylation — the unexplored world of signalling through multiple mono-ubiquitination of Smad3, EMBO J. 30 (2011)
polyubiquitin beyond Lys48 and Lys63 linkages, nature reviews, Mol. Cell. Biol. 13 4777–4789.
(2012) 508–523. [38] E. Maspero, E. Valentini, S. Mari, V. Cecatiello, P. Soffientini, S. Pasqualato,
[9] S.A. Abdul Rehman, Y.A. Kristariyanto, S.Y. Choi, P.J. Nkosi, S. Weidlich, K. Labib, S. Polo, Structure of a ubiquitin-loaded HECT ligase reveals the molecular basis for
K. Hofmann, Y. Kulathu, MINDY-1 is a member of an evolutionarily conserved and catalytic priming, Nat. Struct. Mol. Biol. 20 (2013) 696–701.
structurally distinct new family of deubiquitinating enzymes, Mol. Cell 63 (2016) [39] S.R. Seo, F. Lallemand, N. Ferrand, M. Pessah, S. L'Hoste, J. Camonis, A. Atfi, The
146–155. novel E3 ubiquitin ligase Tiul1 associates with TGIF to target Smad2 for de-
[10] S.M. Nijman, M.P. Luna-Vargas, A. Velds, T.R. Brummelkamp, A.M. Dirac, gradation, EMBO J. 23 (2004) 3780–3792.
T.K. Sixma, R. Bernards, A genomic and functional inventory of deubiquitinating [40] D. Wotton, R.S. Lo, L.-A.C. Swaby, J. Massagué, Multiple modes of repression by
enzymes, Cell 123 (2005) 773–786. the Smad transcriptional corepressor TGIF, J. Biol. Chem. 274 (1999)

477
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

37105–37110. A. Philip, Identification of CD109 as part of the TGF-beta receptor system in


[41] C. Chen, Z. Zhou, J.S. Ross, W. Zhou, J.T. Dong, The amplified WWP1 gene is a human keratinocytes, FASEB J. 20 (2006) 1525–1527.
potential molecular target in breast cancer, Int. J. Cancer 121 (2007) 80–87. [68] H. Fukasawa, T. Yamamoto, Y. Fujigaki, T. Misaki, N. Ohashi, T. Takayama,
[42] C. Chen, Z. Zhou, C.E. Sheehan, E. Slodkowska, C.B. Sheehan, A. Boguniewicz, S. Suzuki, S. Mugiya, T. Oda, C. Uchida, Reduction of transforming growth factor-β
J.S. Ross, Overexpression of WWP1 is associated with the estrogen receptor and type II receptor is caused by the enhanced ubiquitin-dependent degradation in
insulin-like growth factor receptor 1 in breast carcinoma, Int. J. Cancer 124 (2009) human renal cell carcinoma, Int. J. Cancer 127 (2010) 1517–1525.
2829–2836. [69] M. Fukuchi, Y. Fukai, N. Masuda, T. Miyazaki, M. Nakajima, M. Sohda, R. Manda,
[43] C. Chen, X. Sun, P. Guo, X. Dong, P. Sethi, W. Zhou, Z. Zhou, J. Petros, H. Frierson, K. Tsukada, H. Kato, H. Kuwano, High-level expression of the Smad ubiquitin li-
R. Vessella, Ubiquitin E3 ligase WWP1 as an oncogenic factor in human prostate gase Smurf2 correlates with poor prognosis in patients with esophageal squamous
cancer, Oncogene 26 (2007) 2386–2394. cell carcinoma, Cancer Res. 62 (2002) 7162–7165.
[44] T. Courivaud, N. Ferrand, A. Elkhattouti, S. Kumar, L. Levy, O. Ferrigno, A. Atfi, [70] F. Lallemand, S.R. Seo, N. Ferrand, M. Pessah, S. L'Hoste, G. Rawadi, S. Roman-
C. Prunier, Functional characterization of a WWP1/Tiul1 tumor-derived mutant Roman, J. Camonis, A. Atfi, AIP4 restricts transforming growth factor-β signaling
reveals a paradigm of its constitutive activation in human cancer, J. Biol. Chem. through a ubiquitination-independent mechanism, J. Biol. Chem. 280 (2005)
290 (2015) 21007–21018. 27645–27653.
[45] S. Wiesner, A.A. Ogunjimi, H.-R. Wang, D. Rotin, F. Sicheri, J.L. Wrana, [71] Y. Bai, C. Yang, K. Hu, C. Elly, Y.C. Liu, Itch E3 ligase-mediated regulation of TGF-
J.D. Forman-Kay, Autoinhibition of the HECT-type ubiquitin ligase Smurf2 beta signaling by modulating smad2 phosphorylation, Mol. Cell 15 (2004)
through its C2 domain, Cell 130 (2007) 651–662. 825–831.
[46] G. Kuratomi, A. Komuro, G. Kouichiro, M. Shinozaki, K. Miyazawa, K. Miyazono, [72] S.H. Park, E.H. Jung, G.Y. Kim, B.C. Kim, J.H. Lim, C.H. Woo, Itch E3 ubiquitin
T. Imamura, NEDD4-2 (neural precursor cell expressed, developmentally down- ligase positively regulates TGF-beta signaling to EMT via Smad7 ubiquitination,
regulated 4-2) negatively regulates TGF-β (transforming growth factor-β) signal- Mol. Cell 38 (2015) 20–25.
ling by inducing ubiquitin-mediated degradation of Smad2 and TGF-β type I re- [73] Y. Nagano, K.J. Mavrakis, K.L. Lee, T. Fujii, D. Koinuma, H. Sase, K. Yuki,
ceptor, Biochem. J. 386 (2005) 461–470. K. Isogaya, M. Saitoh, T. Imamura, V. Episkopou, K. Miyazono, K. Miyazawa,
[47] M. Fukuchi, T. Imamura, T. Chiba, T. Ebisawa, M. Kawabata, K. Tanaka, Arkadia induces degradation of SnoN and c-Ski to enhance transforming growth
K. Miyazono, Ligand-dependent degradation of Smad3 by a ubiquitin ligase factor-beta signaling, J. Biol. Chem. 282 (2007) 20492–20501.
complex of ROC1 and associated proteins, Mol. Biol. Cell 12 (2001) 1431–1443. [74] D. Koinuma, M. Shinozaki, A. Komuro, K. Goto, M. Saitoh, A. Hanyu, M. Ebina,
[48] J.D. Laney, M. Hochstrasser, Substrate targeting in the ubiquitin system, Cell 97 T. Nukiwa, K. Miyazawa, T. Imamura, K. Miyazono, Arkadia amplifies TGF-beta
(1999) 427–430. superfamily signalling through degradation of Smad7, EMBO J. 22 (2003)
[49] H. Xin, X. Xu, L. Li, H. Ning, Y. Rong, Y. Shang, Y. Wang, X.-Y. Fu, Z. Chang, CHIP 6458–6470.
controls the sensitivity of transforming growth factor-β signaling by modulating [76] L. Levy, M. Howell, D. Das, S. Harkin, V. Episkopou, C.S. Hill, Arkadia activates
the basal level of Smad3 through ubiquitin-mediated degradation, J. Biol. Chem. Smad3/Smad4-dependent transcription by triggering signal-induced SnoN de-
280 (2005) 20842–20850. gradation, Mol. Cell. Biol. 27 (2007) 6068–6083.
[50] M. Wan, Y. Tang, E.M. Tytler, C. Lu, B. Jin, S.M. Vickers, L. Yang, X. Shi, X. Cao, [77] K.J. Mavrakis, R.L. Andrew, K.L. Lee, C. Petropoulou, J.E. Dixon, N. Navaratnam,
Smad4 protein stability is regulated by ubiquitin ligase SCFβ-TrCP1, J. Biol. Chem. D.P. Norris, V. Episkopou, Arkadia enhances Nodal/TGF-β signaling by coupling
279 (2004) 14484–14487. phospho-Smad2/3 activity and turnover, PLoS Biol. 5 (2007) e67.
[51] M. Wan, X. Cao, Y. Wu, S. Bai, L. Wu, X. Shi, N. Wang, X. Cao, Jab1 antagonizes [78] L. Levy, M. Howell, D. Das, S. Harkin, V. Episkopou, C.S. Hill, Arkadia activates
TGF-β signaling by inducing Smad4 degradation, EMBO Rep. 3 (2002) 171–176. Smad3/Smad4-dependent transcription by triggering signal-induced SnoN de-
[52] M. Schutte, R.H. Hruban, L. Hedrick, K.R. Cho, G.M. Nadasdy, C.L. Weinstein, gradation, Mol. Cell. Biol. 27 (2007) 6068–6083.
G.S. Bova, W.B. Isaacs, P. Cairns, H. Nawroz, D. Sidransky, R.A. Casero, [79] S. Bonni, H.R. Wang, C.G. Causing, P. Kavsak, S.L. Stroschein, K. Luo, J.L. Wrana,
P.S. Meltzer, S.A. Hahn, S.E. Kern, Gene in various tumor types, Cancer Res. 56 TGF-beta induces assembly of a Smad2-Smurf2 ubiquitin ligase complex that
(1996) 2527–2530. targets SnoN for degradation, Nat. Cell Biol. 3 (2001) 587–595.
[53] T. Sjöblom, S. Jones, L.D. Wood, D.W. Parsons, J. Lin, T.D. Barber, D. Mandelker, [80] S.L. Stroschein, S. Bonni, J.L. Wrana, K. Luo, Smad3 recruits the anaphase-pro-
R.J. Leary, J. Ptak, N. Silliman, S. Szabo, P. Buckhaults, C. Farrell, P. Meeh, moting complex for ubiquitination and degradation of SnoN, Genes Dev. 15 (2001)
S.D. Markowitz, J. Willis, D. Dawson, J.K.V. Willson, A.F. Gazdar, J. Hartigan, 2822–2836.
L. Wu, C. Liu, G. Parmigiani, B.H. Park, K.E. Bachman, N. Papadopoulos, [81] A. Mizutani, M. Saitoh, T. Imamura, K. Miyazawa, K. Miyazono, Arkadia com-
B. Vogelstein, K.W. Kinzler, V.E. Velculescu, The consensus coding sequences of plexes with clathrin adaptor AP2 and regulates EGF signalling, J. Biochem. 148
human breast and colorectal cancers, Science 314 (2006) 268–274. (2010) 733–741.
[54] A. Moren, U. Hellman, Y. Inada, T. Imamura, C.H. Heldin, A. Moustakas, [82] V. Sharma, A.G. Antonacopoulou, S. Tanaka, A.A. Panoutsopoulos, V. Bravou,
Differential ubiquitination defines the functional status of the tumor suppressor H.P. Kalofonos, V. Episkopou, Enhancement of TGF-β signaling responses by the
Smad4, J. Biol. Chem. 278 (2003) 33571–33582. E3 ubiquitin ligase Arkadia provides tumor suppression in colorectal cancer,
[57] S. Dupont, A. Mamidi, M. Cordenonsi, M. Montagner, L. Zacchigna, M. Adorno, Cancer Res. 71 (2011) 6438–6449.
G. Martello, M.J. Stinchfield, S. Soligo, L. Morsut, FAM/USP9x, a deubiquitinating [83] M.A. Briones-Orta, L. Levy, C.D. Madsen, D. Das, Y. Erker, E. Sahai, C.S. Hill,
enzyme essential for TGFβ signaling, controls Smad4 monoubiquitination, Cell Arkadia regulates tumor metastasis by modulation of the TGF-β pathway, Cancer
136 (2009) 123–135. Res. 73 (2013) 1800–1810.
[58] H. Hayashi, S. Abdollah, Y. Qiu, J. Cai, Y.-Y. Xu, B.W. Grinnell, M.A. Richardson, [84] L. Zhang, H. Huang, F. Zhou, J. Schimmel, C.G. Pardo, T. Zhang, T.S. Barakat, K.-
J.N. Topper, M.A. Gimbrone, J.L. Wrana, The MAD-related protein Smad7 as- A. Sheppard, C. Mickanin, J.A. Porter, RNF12 controls embryonic stem cell fate
sociates with the TGFβ receptor and functions as an antagonist of TGFβ signaling, and morphogenesis in zebrafish embryos by targeting Smad7 for degradation, Mol.
Cell 89 (1997) 1165–1173. Cell 46 (2012) 650–661.
[59] T. Imamura, M. Takase, A. Nishihara, E. Oeda, J.-I. Hanai, M. Kawabata, [85] L. Zhang, F. Zhou, Y. Drabsch, R. Gao, B.E. Snaar-Jagalska, C. Mickanin, H. Huang,
K. Miyazono, Smad6 inhibits signalling by the TGF-β superfamily, Nature 389 K.A. Sheppard, J.A. Porter, C.X. Lu, P. ten Dijke, USP4 is regulated by AKT
(1997) 622–626. phosphorylation and directly deubiquitylates TGF-beta type I receptor, Nat. Cell
[60] A. Nakao, M. Afrakhte, A. Morn, T. Nakayama, J.L. Christian, R. Heuchel, S. Itoh, Biol. 14 (2012) 717–726.
M. Kawabata, N.-E. Heldin, C.-H. Heldin, Identification of Smad7, a TGFβ-in- [86] M.A. Al-Salihi, L. Herhaus, T. Macartney, G.P. Sapkota, USP11 augments TGFbeta
ducible antagonist of TGF-β signalling, Nature 389 (1997) 631–635. signalling by deubiquitylating ALK5, Open Biology 2 (2012) 120063.
[61] A. Hata, G. Lagna, J. Massagué, A. Hemmati-Brivanlou, Smad6 inhibits BMP/ [87] P.J. Eichhorn, L. Rodon, A. Gonzalez-Junca, A. Dirac, M. Gili, E. Martinez-Saez,
Smad1 signaling by specifically competing with the Smad4 tumor suppressor, C. Aura, I. Barba, V. Peg, A. Prat, I. Cuartas, J. Jimenez, D. Garcia-Dorado,
Genes Dev. 12 (1998) 186–197. J. Sahuquillo, R. Bernards, J. Baselga, J. Seoane, USP15 stabilizes TGF-beta re-
[62] S.J. Wicks, K. Haros, M. Maillard, L. Song, R.E. Cohen, P.T. Dijke, A. Chantry, The ceptor I and promotes oncogenesis through the activation of TGF-beta signaling in
deubiquitinating enzyme UCH37 interacts with Smads and regulates TGF-beta glioblastoma, Nat. Med. 18 (2012) 429–435.
signalling, Oncogene 24 (2005) 8080–8084. [88] P.V. Iyengar, P. Jaynes, L. Rodon, D. Lama, K.P. Law, Y.P. Lim, C. Verma,
[63] S. Wiesner, A.A. Ogunjimi, H.R. Wang, D. Rotin, F. Sicheri, J.L. Wrana, J. Seoane, P.J. Eichhorn, USP15 regulates SMURF2 kinetics through C-lobe
J.D. Forman-Kay, Autoinhibition of the HECT-type ubiquitin ligase Smurf2 mediated deubiquitination, Sci Rep 5 (2015) 14733.
through its C2 domain, Cell 130 (2007) 651–662. [89] W.T. Liu, K.Y. Huang, M.C. Lu, H.L. Huang, C.Y. Chen, Y.L. Cheng, H.C. Yu,
[64] A. Komuro, T. Imamura, M. Saitoh, Y. Yoshida, T. Yamori, K. Miyazono, S.Q. Liu, N.S. Lai, H.B. Huang, TGF-beta upregulates the translation of USP15 via
K. Miyazawa, Negative regulation of transforming growth factor-β (TGF-β) sig- the PI3K/AKT pathway to promote p53 stability, Oncogene (2016).
naling by WW domain-containing protein 1 (WWP1), Oncogene 23 (2004) [90] M. Inui, A. Manfrin, A. Mamidi, G. Martello, L. Morsut, S. Soligo, E. Enzo, S. Moro,
6914–6923. S. Polo, S. Dupont, M. Cordenonsi, S. Piccolo, USP15 is a deubiquitylating enzyme
[65] T. Ebisawa, M. Fukuchi, G. Murakami, T. Chiba, K. Tanaka, T. Imamura, for receptor-activated SMADs, Nat. Cell Biol. 13 (2011) 1368–1375.
K. Miyazono, Smurf1 interacts with transforming growth factor-beta type I re- [91] P.J.A. Eichhorn, L. Rodon, A. Gonzalez-Junca, A. Dirac, M. Gili, E. Martinez-Saez,
ceptor through Smad7 and induces receptor degradation, J. Biol. Chem. 276 C. Aura, I. Barba, V. Peg, A. Prat, I. Cuartas, J. Jimenez, D. Garcia-Dorado,
(2001) 12477–12480. J. Sahuquillo, R. Bernards, J. Baselga, J. Seoane, USP15 stabilizes TGF-[beta] re-
[66] G. Kuratomi, A. Komuro, K. Goto, M. Shinozaki, K. Miyazawa, K. Miyazono, ceptor I and promotes oncogenesis through the activation of TGF-[beta] signaling
T. Imamura, NEDD4–2 (neural precursor cell expressed, developmentally down- in glioblastoma, Nat. Med. 18 (2012) 429–435.
regulated 4–2) negatively regulates TGF-beta (transforming growth factor-beta) [92] L. Zhang, F. Zhou, Y. Drabsch, R. Gao, B.E. Snaar-Jagalska, C. Mickanin, H. Huang,
signalling by inducing ubiquitin-mediated degradation of Smad2 and TGF-beta K.-A. Sheppard, J.A. Porter, C.X. Lu, USP4 is regulated by AKT phosphorylation
type I receptor, The Biochemical Journal 386 (2005) 461–470. and directly deubiquitylates TGF-[beta] type I receptor, Nat. Cell Biol. 14 (2012)
[67] K.W. Finnson, B.Y. Tam, K. Liu, A. Marcoux, P. Lepage, S. Roy, A.A. Bizet, 717–726.

478
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

[93] M.A. Al-Salihi, L. Herhaus, T. Macartney, G.P. Sapkota, USP11 augments TGFβ W.C. Hahn, KRAS and YAP1 converge to regulate EMT and tumor survival, Cell
signalling by deubiquitylating ALK5, Open Biology 2 (2012) 120063. 158 (2014) 171–184.
[94] S. Kit Leng Lui, P.V. Iyengar, P. Jaynes, Z. Isa, B. Pang, T.Z. Tan, P.J.A. Eichhorn, [122] W. Zhang, Y. Gao, F. Li, X. Tong, Y. Ren, X. Han, S. Yao, F. Long, Z. Yang, H. Fan,
USP26 regulates TGF-beta signaling by deubiquitinating and stabilizing SMAD7, L. Zhang, H. Ji, YAP promotes malignant progression of Lkb1-deficient lung
EMBO Rep. 18 (2017) 797–808. adenocarcinoma through downstream regulation of survivin, Cancer Res. 75
[95] F. Itoh, H. Asao, K. Sugamura, C.H. Heldin, P. ten Dijke, S. Itoh, Promoting bone (2015) 4450–4457.
morphogenetic protein signaling through negative regulation of inhibitory Smads, [123] W. Zhang, N. Nandakumar, Y. Shi, M. Manzano, A. Smith, G. Graham, S. Gupta,
EMBO J. 20 (2001) 4132–4142. E.E. Vietsch, S.Z. Laughlin, M. Wadhwa, M. Chetram, M. Joshi, F. Wang,
[96] N. Tanaka, K. Kaneko, H. Asao, H. Kasai, Y. Endo, T. Fujita, T. Takeshita, B. Kallakury, J. Toretsky, A. Wellstein, C. Yi, Downstream of mutant KRAS, the
K. Sugamura, Possible involvement of a novel STAM-associated molecule “AMSH” transcription regulator YAP is essential for neoplastic progression to pancreatic
in intracellular signal transduction mediated by cytokines, J. Biol. Chem. 274 ductal adenocarcinoma, Sci. Signal. 7 (2014) ra42.
(1999) 19129–19135. [124] H.T. Nguyen, J.M. Kugler, S.M. Cohen, DUB3 deubiquitylating enzymes regulate
[97] J.H. Hurley, Nipped in the bud: how the AMSH MIT domain helps deubiquitinate hippo pathway activity by regulating the stability of ITCH, LATS and AMOT
lysosome-bound cargo, Structure 19 (2011) 1033–1035. proteins, PLoS One 12 (2017) e0169587.
[98] L. Herhaus, M. Al-Salihi, T. Macartney, S. Weidlich, G.P. Sapkota, OTUB1 en- [125] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144
hances TGFβ signalling by inhibiting the ubiquitylation and degradation of active (2011) 646–674.
SMAD2/3, Nat. Commun. 4 (2013). [126] S. Sigismund, S. Confalonieri, A. Ciliberto, S. Polo, G. Scita, P.P. Di Fiore,
[99] S. Nakada, I. Tai, S. Panier, A. Al-Hakim, S.-i. Iemura, Y.-C. Juang, L. O'Donnell, Endocytosis and signaling: cell logistics shape the eukaryotic cell plan, Physiol.
A. Kumakubo, M. Munro, F. Sicheri, Non-canonical inhibition of DNA damage- Rev. 92 (2012) 273–366.
dependent ubiquitination by OTUB1, Nature 466 (2010) 941–946. [127] K. Haglund, I. Dikic, The role of ubiquitylation in receptor endocytosis and en-
[100] Y. Sato, A. Yamagata, S. Goto-Ito, K. Kubota, R. Miyamoto, S. Nakada, S. Fukai, dosomal sorting, J. Cell Sci. 125 (2012) 265–275.
Molecular basis of Lys-63-linked polyubiquitination inhibition by the interaction [128] M.G. Savio, N. Wollscheid, E. Cavallaro, V. Algisi, P.P. Di Fiore, S. Sigismund,
between human deubiquitinating enzyme OTUB1 and ubiquitin-conjugating E. Maspero, S. Polo, USP9X controls EGFR fate by deubiquitinating the endocytic
enzyme UBC13, J. Biol. Chem. 287 (2012) 25860–25868. adaptor Eps15, Current Biology: CB 26 (2016) 173–183.
[101] Y.-C. Juang, M.-C. Landry, M. Sanches, V. Vittal, C.C.Y. Leung, Derek [129] I.M. Meijer, J.E. van Leeuwen, ERBB2 is a target for USP8-mediated deubiquiti-
F. Ceccarelli, A.-Rachele F. Mateo, Jonathan N. Pruneda, D.Y.L. Mao, Rachel nation, Cell. Signal. 23 (2011) 458–467.
K. Szilard, S. Orlicky, M. Munro, Peter S. Brzovic, Rachel E. Klevit, F. Sicheri, [130] S. Niendorf, A. Oksche, A. Kisser, J. Löhler, M. Prinz, H. Schorle, S. Feller,
D. Durocher, OTUB1 Co-opts Lys48-linked ubiquitin recognition to suppress E2 M. Lewitzky, I. Horak, K.-P. Knobeloch, Essential role of ubiquitin-specific pro-
enzyme function, Mol. Cell 45 (2012) 384–397. tease 8 for receptor tyrosine kinase stability and endocytic trafficking in vivo, Mol.
[102] R. Wiener, X. Zhang, T. Wang, C. Wolberger, The mechanism of OTUB1-mediated Cell. Biol. 27 (2007) 5029–5039.
inhibition of ubiquitination, Nature 483 (2012) 618–622. [131] I.M. Meijer, J. Kerperien, A.M. Sotoca, E.J. van Zoelen, J.E. van Leeuwen, The
[103] S. Dupont, L. Zacchigna, M. Cordenonsi, S. Soligo, M. Adorno, M. Rugge, Usp8 deubiquitination enzyme is post-translationally modified by tyrosine and
S. Piccolo, Germ-layer specification and control of cell growth by ectodermin, a serine phosphorylation, Cell. Signal. 25 (2013) 919–930.
Smad4 ubiquitin ligase, Cell 121 (2005) 87–99. [132] T. Obsil, V. Obsilova, Structural Basis of 14-3-3 Protein Functions, Seminars in
[104] Z. Meng, T. Moroishi, K.L. Guan, Mechanisms of hippo pathway regulation, Genes Cell & Developmental Biology, Elsevier, 2011, pp. 663–672.
Dev. 30 (2016) 1–17. [133] X. Wu, L. Yen, L. Irwin, C. Sweeney, K.L. Carraway, Stabilization of the E3 ubi-
[105] F. Zanconato, M. Cordenonsi, S. Piccolo, YAP/TAZ at the roots of cancer, Cancer quitin ligase Nrdp1 by the deubiquitinating enzyme USP8, Mol. Cell. Biol. 24
Cell 29 (2016) 783–803. (2004) 7748–7757.
[106] T. Moroishi, C.G. Hansen, K.L. Guan, The emerging roles of YAP and TAZ in [134] Z. Liu, S.M. Zanata, J. Kim, M.A. Peterson, D. Di Vizio, L.R. Chirieac, S. Pyne,
cancer, Nat. Rev. Cancer 15 (2015) 73–79. M. Agostini, M.R. Freeman, M. Loda, The ubiquitin-specific protease USP2a pre-
[107] S. Piccolo, S. Dupont, M. Cordenonsi, The biology of YAP/TAZ: hippo signaling vents endocytosis-mediated EGFR degradation, Oncogene 32 (2013) 1660–1669.
and beyond, Physiol. Rev. 94 (2014) 1287–1312. [135] J. Kim, W.-J. Kim, Z. Liu, M. Loda, M.R. Freeman, The ubiquitin-specific protease
[108] X. Hong, H.T. Nguyen, Q. Chen, R. Zhang, Z. Hagman, P.M. Voorhoeve, USP2a enhances tumor progression by targeting cyclin A1 in bladder cancer, Cell
S.M. Cohen, Opposing activities of the Ras and hippo pathways converge on Cycle 11 (2012) 1123–1130.
regulation of YAP protein turnover, EMBO J. 33 (2014) 2447–2457. [136] B. Sehat, S. Andersson, R. Vasilcanu, L. Girnita, O. Larsson, Role of ubiquitination
[109] B. Yeung, K.C. Ho, X. Yang, WWP1 E3 ligase targets LATS1 for ubiquitin-mediated in IGF-1 receptor signaling and degradation, PLoS One 2 (2007) e340.
degradation in breast cancer cells, PLoS One 8 (2013) e61027. [137] G. Levkowitz, H. Waterman, S.A. Ettenberg, M. Katz, A.Y. Tsygankov, I. Alroy,
[110] S.J. Bae, M. Kim, S.H. Kim, Y.E. Kwon, J.H. Lee, J. Kim, C.H. Chung, W.J. Lee, S. Lavi, K. Iwai, Y. Reiss, A. Ciechanover, Ubiquitin ligase activity and tyrosine
J.H. Seol, NEDD4 controls intestinal stem cell homeostasis by regulating the hippo phosphorylation underlie suppression of growth factor signaling by c-Cbl/Sli-1,
signalling pathway, Nat. Commun. 6 (2015) 6314. Mol. Cell 4 (1999) 1029–1040.
[111] K.C. Ho, Z. Zhou, Y.M. She, A. Chun, T.D. Cyr, X. Yang, Itch E3 ubiquitin ligase [138] H. Waterman, M. Katz, C. Rubin, K. Shtiegman, S. Lavi, A. Elson, T. Jovin,
regulates large tumor suppressor 1 stability [corrected], Proc. Natl. Acad. Sci. U. S. Y. Yarden, A mutant EGF-receptor defective in ubiquitylation and endocytosis
A. 108 (2011) 4870–4875. unveils a role for Grb2 in negative signaling, EMBO J. 21 (2002) 303–313.
[112] Z. Salah, G. Melino, R.I. Aqeilan, Negative regulation of the hippo pathway by E3 [139] H. Dou, L. Buetow, A. Hock, G.J. Sibbet, K.H. Vousden, D.T. Huang, Structural
ubiquitin ligase ITCH is sufficient to promote tumorigenicity, Cancer Res. 71 basis for autoinhibition and phosphorylation-dependent activation of c-Cbl, Nat.
(2011) 2010–2020. Struct. Mol. Biol. 19 (2012) 184–192.
[113] R.J. Ingham, G. Gish, T. Pawson, The Nedd4 family of E3 ubiquitin ligases: [140] G. Christofori, Split personalities: the agonistic antagonist Sprouty, Nat. Cell Biol.
functional diversity within a common modular architecture, Oncogene 23 (2004) 5 (2003) 377–379.
1972–1984. [141] C. Rubin, V. Litvak, H. Medvedovsky, Y. Zwang, S. Lev, Y. Yarden, Sprouty fine-
[114] B. Ma, Y. Chen, L. Chen, H. Cheng, C. Mu, J. Li, R. Gao, C. Zhou, L. Cao, J. Liu, tunes EGF signaling through interlinked positive and negative feedback loops,
Y. Zhu, Q. Chen, S. Wu, Hypoxia regulates hippo signalling through the SIAH2 Curr. Biol. 13 (2003) 297–307.
ubiquitin E3 ligase, Nat. Cell Biol. 17 (2015) 95–103. [142] G.R. Guy, R.A. Jackson, P. Yusoff, S.Y. Chow, Sprouty proteins: modified mod-
[115] L. Lignitto, A. Arcella, M. Sepe, L. Rinaldi, R. Delle Donne, A. Gallo, E. Stefan, ulators, matchmakers or missing links? J. Endocrinol. 203 (2009) 191–202.
V.A. Bachmann, M.A. Oliva, C. Tiziana Storlazzi, A. L'Abbate, A. Brunetti, [143] H. Hanafusa, S. Torii, T. Yasunaga, E. Nishida, Sprouty1 and Sprouty2 provide a
S. Gargiulo, M. Gramanzini, L. Insabato, C. Garbi, M.E. Gottesman, A. Feliciello, control mechanism for the Ras/MAPK signalling pathway, Nat. Cell Biol. 4 (2002)
Proteolysis of MOB1 by the ubiquitin ligase praja2 attenuates hippo signalling and 850–858.
supports glioblastoma growth, Nat. Commun. 4 (2013) 1822. [144] M. Tsang, I.B. Dawid, Promotion and attenuation of FGF signaling through the
[116] B. Zhao, L. Li, Q. Lu, L.H. Wang, C.Y. Liu, Q. Lei, K.L. Guan, Angiomotin is a novel Ras-MAPK pathway, Sci. STKE 2004 (2004) e17.
hippo pathway component that inhibits YAP oncoprotein, Genes Dev. 25 (2011) [145] A. Sasaki, T. Taketomi, R. Kato, K. Saeki, A. Nonami, M. Sasaki, M. Kuriyama,
51–63. N. Saito, M. Shibuya, A. Yoshimura, Mammalian Sprouty4 suppresses Ras-in-
[117] J.J. Adler, D.E. Johnson, B.L. Heller, L.R. Bringman, W.P. Ranahan, M.D. Conwell, dependent ERK activation by binding to Raf1, Nat. Cell Biol. 5 (2003) 427–432.
Y. Sun, A. Hudmon, C.D. Wells, Serum deprivation inhibits the transcriptional co- [146] A.B. Keeton, E.A. Salter, G.A. Piazza, The RAS–effector interaction as a drug target,
activator YAP and cell growth via phosphorylation of the 130-kDa isoform of Cancer Res. (2017).
angiomotin by the LATS1/2 protein kinases, Proc. Natl. Acad. Sci. U. S. A. 110 [147] N. Jura, E. Scotto-Lavino, A. Sobczyk, D. Bar-Sagi, Differential modification of Ras
(2013) 17368–17373. proteins by ubiquitination, Mol. Cell 21 (2006) 679–687.
[118] M. Kim, M. Kim, S.J. Park, C. Lee, D.S. Lim, Role of angiomotin-like 2 mono- [148] S.-E. Kim, J.-Y. Yoon, W.-J. Jeong, S.-H. Jeon, Y. Park, J.-B. Yoon, Y.N. Park,
ubiquitination on YAP inhibition, EMBO Rep. 17 (2016) 64–78. H. Kim, K.-Y. Choi, H-Ras is degraded by Wnt/β-catenin signaling via β-TrCP-
[119] H.T. Nguyen, X. Hong, S. Tan, Q. Chen, L. Chan, M. Fivaz, S.M. Cohen, mediated polyubiquitylation, J. Cell Sci. 122 (2009) 842–848.
P.M. Voorhoeve, Viral small T oncoproteins transform cells by alleviating hippo- [149] S. Shukla, U.S. Allam, A. Ahsan, G. Chen, P.M. Krishnamurthy, K. Marsh,
pathway-mediated inhibition of the YAP proto-oncogene, Cell Rep. 8 (2014) M. Rumschlag, S. Shankar, C. Whitehead, M. Schipper, V. Basrur,
707–713. D.R. Southworth, A.M. Chinnaiyan, A. Rehemtulla, D.G. Beer, T.S. Lawrence,
[120] D. Shao, P. Zhai, D.P. Del Re, S. Sciarretta, N. Yabuta, H. Nojima, D.S. Lim, D. Pan, M.K. Nyati, D. Ray, KRAS protein stability is regulated through SMURF2: UBCH5
J. Sadoshima, A functional interaction between hippo-YAP signalling and FoxO1 complex-mediated beta-TrCP1 degradation, Neoplasia 16 (2014) 115–128.
mediates the oxidative stress response, Nat. Commun. 5 (2014) 3315. [150] H. Yan, M. Jahanshahi, E.A. Horvath, H.Y. Liu, C.M. Pfleger, Rabex-5 ubiquitin
[121] D.D. Shao, W. Xue, E.B. Krall, A. Bhutkar, F. Piccioni, X. Wang, A.C. Schinzel, ligase activity restricts Ras signaling to establish pathway homeostasis in droso-
S. Sood, J. Rosenbluh, J.W. Kim, Y. Zwang, T.M. Roberts, D.E. Root, T. Jacks, phila, Current Biology: CB 20 (2010) 1378–1382.

479
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

[151] L. Xu, V. Lubkov, L.J. Taylor, D. Bar-Sagi, Feedback regulation of Ras signaling by ubiquitination and activation, Science 325 (2009) 1134–1138.
Rabex-5-mediated ubiquitination, Current Biology: CB 20 (2010) 1372–1377. [178] C.-D. Fan, M.A. Lum, C. Xu, J.D. Black, X. Wang, Ubiquitin-dependent regulation
[152] R. Baker, E.M. Wilkerson, K. Sumita, D.G. Isom, A.T. Sasaki, H.G. Dohlman, of phospho-AKT dynamics by the ubiquitin E3 ligase, NEDD4-1, in the insulin-like
S.L. Campbell, Differences in the regulation of K-Ras and H-Ras isoforms by growth factor-1 response, J. Biol. Chem. 288 (2013) 1674–1684.
monoubiquitination, J. Biol. Chem. 288 (2013) 36856–36862. [179] W. Li, C. Peng, M.H. Lee, D. Lim, F. Zhu, Y. Fu, G. Yang, Y. Sheng, L. Xiao, X. Dong,
[153] A. Laine, Ubiquitin chains in the ladder of MAPK signaling, Sci. Signal. 2005 W. Ma, A.M. Bode, Y. Cao, Z. Dong, TRAF4 is a critical molecule for Akt activation
(2005) re5-re5. in lung cancer, Cancer Res. 73 (2013) 6938–6950.
[154] S.A. Matheny, C. Chen, R.L. Kortum, G.L. Razidlo, R.E. Lewis, M.A. White, Ras [180] C.-H. Chan, C.-F. Li, W.-L. Yang, Y. Gao, S.-W. Lee, Z. Feng, H.-Y. Huang, K.K. Tsai,
regulates assembly of mitogenic signalling complexes through the effector protein L.G. Flores, Y. Shao, The Skp2-SCF E3 ligase regulates Akt ubiquitination, glyco-
IMP, Nature 427 (2004) 256–260. lysis, herceptin sensitivity, and tumorigenesis, Cell 149 (2012) 1098–1111.
[155] S.D. Hayes, H. Liu, E. MacDonald, C.M. Sanderson, J.M. Coulson, M.J. Clague, [181] J.D. Carpten, A.L. Faber, C. Horn, G.P. Donoho, S.L. Briggs, C.M. Robbins,
S. Urbé, Direct and indirect control of mitogen-activated protein kinase pathway- G. Hostetter, S. Boguslawski, T.Y. Moses, S. Savage, M. Uhlik, A. Lin, J. Du,
associated components, BRAP/IMP E3 ubiquitin ligase and CRAF/RAF1 kinase, by Y.W. Qian, D.J. Zeckner, G. Tucker-Kellogg, J. Touchman, K. Patel, S. Mousses,
the deubiquitylating enzyme USP15, J. Biol. Chem. 287 (2012) 43007–43018. M. Bittner, R. Schevitz, M.H. Lai, K.L. Blanchard, J.E. Thomas, A transforming
[156] E.R. Jang, P. Shi, J. Bryant, J. Chen, V. Dukhande, M.S. Gentry, H. Jang, mutation in the pleckstrin homology domain of AKT1 in cancer, Nature 448
M. Jeoung, E. Galperin, HUWE1 is a molecular link controlling RAF-1 activity (2007) 439–444.
supported by the Shoc2 scaffold, Mol. Cell. Biol. 34 (2014) 3579–3593. [182] J.H. Lim, H. Jono, K. Komatsu, C.-H. Woo, J. Lee, M. Miyata, T. Matsuno, X. Xu,
[157] T. Dogan, G.S. Harms, M. Hekman, C. Karreman, T.K. Oberoi, E.S. Alnemri, Y. Huang, W. Zhang, CYLD negatively regulates transforming growth factor-β-
U.R. Rapp, K. Rajalingam, X-linked and cellular IAPs modulate the stability of C- signalling via deubiquitinating Akt, Nat. Commun. 3 (2012) 771.
RAF kinase and cell motility, Nat. Cell Biol. 10 (2008) 1447–1455. [183] W.-L. Yang, G. Jin, C.-F. Li, Y.S. Jeong, A. Moten, D. Xu, Z. Feng, W. Chen, Z. Cai,
[158] T.K. Oberoi-Khanuja, C. Karreman, S. Larisch, U.R. Rapp, K. Rajalingam, Role of B. Darnay, Cycles of ubiquitination and deubiquitination critically regulate growth
melanoma inhibitor of apoptosis (ML-IAP) protein, a member of the baculoviral factor-mediated activation of Akt signaling, Sci. Signal. 6 (2013).
IAP repeat (BIR) domain family, in the regulation of C-RAF kinase and cell mi- [184] E. Guven-Maiorov, O. Keskin, A. Gursoy, R. Nussinov, A structural view of nega-
gration, J. Biol. Chem. 287 (2012) 28445–28455. tive regulation of the toll-like receptor-mediated inflammatory pathway, Biophys.
[159] O. Grbovic, A. Basso, A. Sawai, Q. Ye, P. Friedlander, D. Solit, N. Rosen, V600E B- J. 109 (2015) 1214–1226.
Raf requires the Hsp90 chaperone for stability and is degraded in response to [185] M. Li, C.L. Brooks, N. Kon, W. Gu, A dynamic role of HAUSP in the p53-Mdm2
Hsp90 inhibitors, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 57–62. pathway, Mol. Cell 13 (2004) 879–886.
[160] T.W. Schulte, M.V. Blagosklonny, C. Ingui, L. Neckers, Disruption of the Raf-1- [186] T. Xiang, A. Ohashi, Y. Huang, T.K. Pandita, T. Ludwig, S.N. Powell, Q. Yang,
Hsp90 molecular complex results in destabilization of Raf-1 and loss of Raf-1-Ras Negative regulation of AKT activation by BRCA1, Cancer Res. 68 (2008)
association, J. Biol. Chem. 270 (1995) 24585–24588. 10040–10044.
[161] T.W. Schulte, W.G. An, L.M. Neckers, Geldanamycin-induced destabilization of [187] S. Bae, S.Y. Kim, J.H. Jung, Y. Yoon, H.J. Cha, H. Lee, K. Kim, J. Kim, I.S. An,
Raf-1 involves the proteasome, Biochem. Biophys. Res. Commun. 239 (1997) J. Kim, H.D. Um, I.C. Park, S.J. Lee, S.Y. Nam, Y.W. Jin, J.H. Lee, S. An, Akt is
655–659. negatively regulated by the MULAN E3 ligase, Cell Res. 22 (2012) 873–885.
[162] S.-W. Hong, D.-H. Jin, J.-S. Shin, J.-H. Moon, Y.-S. Na, K.-A. Jung, S.-M. Kim, [188] C.A. Dickey, J. Koren, Y.J. Zhang, Y.F. Xu, U.K. Jinwal, M.J. Birnbaum, B. Monks,
J.C. Kim, K.-p. Kim, Y.S. Hong, Ring finger protein 149 is an E3 ubiquitin ligase M. Sun, J.Q. Cheng, C. Patterson, R.M. Bailey, J. Dunmore, S. Soresh, C. Leon,
active on wild-type v-Raf murine sarcoma viral oncogene homolog B1 (BRAF), J. D. Morgan, L. Petrucelli, Akt and CHIP coregulate tau degradation through co-
Biol. Chem. 287 (2012) 24017–24025. ordinated interactions, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 3622–3627.
[163] J. Du, J. Zeng, X. Ou, X. Ren, S. Cai, Methylglyoxal downregulates Raf-1 protein [189] F. Suizu, Y. Hiramuki, F. Okumura, M. Matsuda, A.J. Okumura, N. Hirata,
through a ubiquitination-mediated mechanism, Int. J. Biochem. Cell Biol. 38 M. Narita, T. Kohno, J. Yokota, M. Bohgaki, C. Obuse, S. Hatakeyama, T. Obata,
(2006) 1084–1091. M. Noguchi, The E3 ligase TTC3 facilitates ubiquitination and degradation of
[164] Z. Lu, S. Xu, C. Joazeiro, M.H. Cobb, T. Hunter, The PHD domain of MEKK1 acts as phosphorylated Akt, Dev. Cell 17 (2009) 800–810.
an E3 ubiquitin ligase and mediates ubiquitination and degradation of ERK1/2, [190] A. Toker, L.C. Cantley, Signalling through the lipid products of phosphoinositide-
Mol. Cell 9 (2002) 945–956. 3-OH kinase, Nature 387 (1997) 673–676.
[165] L.C. Cantley, B.G. Neel, New insights into tumor suppression: PTEN suppresses [191] P. Rodriguez-Viciana, P.H. Warne, A. Khwaja, B.M. Marte, D. Pappin, P. Das,
tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway, Proc. M.D. Waterfield, A. Ridley, J. Downward, Role of phosphoinositide 3-OH kinase in
Natl. Acad. Sci. 96 (1999) 4240–4245. cell transformation and control of the actin cytoskeleton by Ras, Cell 89 (1997)
[166] M. Cully, H. You, A.J. Levine, T.W. Mak, Beyond PTEN mutations: the PI3K 457–467.
pathway as an integrator of multiple inputs during tumorigenesis, Nat. Rev. [192] L.R. Pearce, D. Komander, D.R. Alessi, The nuts and bolts of AGC protein kinases,
Cancer 6 (2006) 184–192. Nat. Rev. Mol. Cell Biol. 11 (2010) 9–22.
[167] S. Kuchay, S. Duan, E. Schenkein, A. Peschiaroli, A. Saraf, L. Florens, [193] I.Z. Uras, T. List, S.M. Nijman, Ubiquitin-specific protease 4 inhibits mono-ubi-
M.P. Washburn, M. Pagano, FBXL2-and PTPL1-mediated degradation of p110-free quitination of the master growth factor signaling kinase PDK1, PLoS One 7 (2012)
p85β regulatory subunit controls the PI (3) K signalling cascade, Nat. Cell Biol. 15 e31003.
(2013) 472–480. [194] M. Laplante, D.M. Sabatini, mTOR signaling at a glance, J. Cell Sci. 122 (2009)
[168] B. Geering, P. Cutillas, B. Vanhaesebroeck, Regulation of class IA PI3Ks: is there a 3589–3594.
role for monomeric PI3K subunits? Biochem. Soc. Trans. 35 (2007) 199–203. [195] M. Corradetti, K. Guan, Upstream of the mammalian target of rapamycin: do all
[169] H. Ko, C. Kim, S. Lee, J. Song, K. Lee, K. Kim, K. Park, S. Cho, J. Ahn, P42 Ebp1 roads pass through mTOR? Oncogene 25 (2006) 6347–6360.
regulates the proteasomal degradation of the p85 regulatory subunit of PI3K by [196] T.R. Peterson, M. Laplante, C.C. Thoreen, Y. Sancak, S.A. Kang, W.M. Kuehl,
recruiting a chaperone-E3 ligase complex HSP70/CHIP, Cell Death Dis. 5 (2014) N.S. Gray, D.M. Sabatini, DEPTOR is an mTOR inhibitor frequently overexpressed
e1131. in multiple myeloma cells and required for their survival, Cell 137 (2009)
[170] D. Fang, H.-Y. Wang, N. Fang, Y. Altman, C. Elly, Y.-C. Liu, Cbl-b, a RING-type E3 873–886.
ubiquitin ligase, targets phosphatidylinositol 3-kinase for ubiquitination in T cells, [197] Y. Zhao, X. Xiong, Y. Sun, DEPTOR, an mTOR inhibitor, is a physiological sub-
J. Biol. Chem. 276 (2001) 4872–4878. strate of SCF βTrCP E3 ubiquitin ligase and regulates survival and autophagy, Mol.
[171] X. Xu, A. Sarikas, D.C. Dias-Santagata, G. Dolios, P.J. Lafontant, S.-C. Tsai, W. Zhu, Cell 44 (2011) 304–316.
H. Nakajima, H.O. Nakajima, L.J. Field, The CUL7 E3 ubiquitin ligase targets in- [198] D. Gao, H. Inuzuka, M.-K.M. Tan, H. Fukushima, J.W. Locasale, P. Liu, L. Wan,
sulin receptor substrate 1 for ubiquitin-dependent degradation, Mol. Cell 30 B. Zhai, Y.R. Chin, S. Shaik, mTOR drives its own activation via SCF βTrCP-de-
(2008) 403–414. pendent degradation of the mTOR inhibitor DEPTOR, Mol. Cell 44 (2011)
[172] L. Rui, M. Yuan, D. Frantz, S. Shoelson, M.F. White, SOCS-1 and SOCS-3 block 290–303.
insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2, J. Biol. [199] S. Duan, J.R. Skaar, S. Kuchay, A. Toschi, N. Kanarek, Y. Ben-Neriah, M. Pagano,
Chem. 277 (2002) 42394–42398. mTOR generates an auto-amplification loop by triggering the βTrCP-and CK1α-
[173] R. Nakao, K. Hirasaka, J. Goto, K. Ishidoh, C. Yamada, A. Ohno, Y. Okumura, dependent degradation of DEPTOR, Mol. Cell 44 (2011) 317–324.
I. Nonaka, K. Yasutomo, K.M. Baldwin, Ubiquitin ligase Cbl-b is a negative reg- [200] P. Ghosh, M. Wu, H. Zhang, H. Sun, mTORC1 signaling requires proteasomal
ulator for insulin-like growth factor 1 signaling during muscle atrophy caused by function and the involvement of CUL4-DDB1 ubiquitin E3 ligase, Cell Cycle 7
unloading, Mol. Cell. Biol. 29 (2009) 4798–4811. (2008) 373–381.
[174] R. Song, W. Peng, Y. Zhang, F. Lv, H.-K. Wu, J. Guo, Y. Cao, Y. Pi, X. Zhang, L. Jin, [201] S. Hussain, A.L. Feldman, C. Das, S.C. Ziesmer, S.M. Ansell, P.J. Galardy, Ubiquitin
Central role of E3 ubiquitin ligase MG53 in insulin resistance and metabolic dis- hydrolase UCH-L1 destabilizes mTOR complex 1 by antagonizing DDB1-CUL4-
orders, Nature 494 (2013) 375–379. mediated ubiquitination of raptor, Mol. Cell. Biol. 33 (2013) 1188–1197.
[175] T. Fukushima, H. Yoshihara, H. Furuta, H. Kamei, F. Hakuno, J. Luan, C. Duan, [202] B. Wang, Z. Jie, D. Joo, A. Ordureau, P. Liu, W. Gan, J. Guo, J. Zhang, B.J. North,
Y. Saeki, K. Tanaka, S.-I. Iemura, Nedd4-induced monoubiquitination of IRS-2 X. Dai, X. Cheng, X. Bian, L. Zhang, J.W. Harper, S.C. Sun, W. Wei, TRAF2 and
enhances IGF signalling and mitogenic activity, Nat. Commun. 6 (2015). OTUD7B govern a ubiquitin-dependent switch that regulates mTORC2 signalling,
[176] H. Yoshihara, T. Fukushima, F. Hakuno, Y. Saeki, K. Tanaka, A. Ito, M. Yoshida, S.- Nature 545 (2017) 365–369.
I. Iemura, T. Natsume, T. Asano, Insulin/insulin-like growth factor (IGF) stimu- [203] J.-H. Mao, I.-J. Kim, D. Wu, J. Climent, H.C. Kang, R. DelRosario, A. Balmain,
lation abrogates an association between a deubiquitinating enzyme USP7 and FBXW7 targets mTOR for degradation and cooperates with PTEN in tumor sup-
insulin receptor substrates (IRSs) followed by proteasomal degradation of IRSs, pression, Science 321 (2008) 1499–1502.
Biochem. Biophys. Res. Commun. 423 (2012) 122–127. [204] P. Agrawal, Y.-T. Chen, B. Schilling, B.W. Gibson, R.E. Hughes, Ubiquitin-specific
[177] W.-L. Yang, J. Wang, C.-H. Chan, S.-W. Lee, A.D. Campos, B. Lamothe, L. Hur, peptidase 9, X-linked (USP9X) modulates activity of mammalian target of rapa-
B.C. Grabiner, X. Lin, B.G. Darnay, The E3 ligase TRAF6 regulates Akt mycin (mTOR), J. Biol. Chem. 287 (2012) 21164–21175.

480
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

[205] G. Panasyuk, I. Nemazanyy, V. Filonenko, I. Gout, Ribosomal protein S6 kinase 1 [231] C. Dominguez-Brauer, R. Khatun, A.J. Elia, K.L. Thu, P. Ramachandran,
interacts with and is ubiquitinated by ubiquitin ligase ROC1, Biochem. Biophys. S.P. Baniasadi, Z. Hao, L.D. Jones, J. Haight, Y. Sheng, T.W. Mak, E3 ubiquitin
Res. Commun. 369 (2008) 339–343. ligase Mule targets beta-catenin under conditions of hyperactive Wnt signaling,
[206] S. Hussain, O. Foreman, S. Perkins, T.E. Witzig, R. Miles, J. Van Deursen, Proc. Natl. Acad. Sci. U. S. A. 114 (2017) E1148–E1157.
P.J. Galardy, The de-ubiquitinase UCH-L1 is an oncogene that drives the devel- [232] S. Angers, C.J. Thorpe, T.L. Biechele, S.J. Goldenberg, N. Zheng, M.J. MacCoss,
opment of lymphoma in vivo by deregulating PHLPP1 and Akt signaling, R.T. Moon, The KLHL12-Cullin-3 ubiquitin ligase negatively regulates the Wnt-
Leukemia 24 (2010) 1641–1655. beta-catenin pathway by targeting dishevelled for degradation, Nat. Cell Biol. 8
[207] I. Vivanco, D. Rohle, M. Versele, A. Iwanami, D. Kuga, B. Oldrini, K. Tanaka, (2006) 348–357.
J. Dang, S. Kubek, N. Palaskas, The phosphatase and tensin homolog regulates [233] W. Wei, M. Li, J. Wang, F. Nie, L. Li, The E3 ubiquitin ligase ITCH negatively
epidermal growth factor receptor (EGFR) inhibitor response by targeting EGFR for regulates canonical Wnt signaling by targeting dishevelled protein, Mol. Cell. Biol.
degradation, Proc. Natl. Acad. Sci. 107 (2010) 6459–6464. 32 (2012) 3903–3912.
[208] T. Tolkacheva, M. Boddapati, A. Sanfiz, K. Tsuchida, A.C. Kimmelman, A.M. Chan, [234] Y. Ding, Y. Zhang, C. Xu, Q.H. Tao, Y.G. Chen, HECT domain-containing E3 ubi-
Regulation of PTEN binding to MAGI-2 by two putative phosphorylation sites at quitin ligase NEDD4L negatively regulates Wnt signaling by targeting dishevelled
threonine 382 and 383, Cancer Res. 61 (2001) 4985–4989. for proteasomal degradation, J. Biol. Chem. 288 (2013) 8289–8298.
[209] E.-M. Duerr, B. Rollbrocker, Y. Hayashi, N. Peters, B. Meyer-Puttlitz, D.N. Louis, [235] D.V. Tauriello, A. Haegebarth, I. Kuper, M.J. Edelmann, M. Henraat,
J. Schramm, O.D. Wiestler, R. Parsons, C. Eng, PTEN mutations in gliomas and M.R. Canninga-van Dijk, B.M. Kessler, H. Clevers, M.M. Maurice, Loss of the tumor
glioneuronal tumors, Oncogene 16 (1998). suppressor CYLD enhances Wnt/beta-catenin signaling through K63-linked ubi-
[210] L.C. Trotman, X. Wang, A. Alimonti, Z. Chen, J. Teruya-Feldstein, H. Yang, quitination of Dvl, Mol. Cell 37 (2010) 607–619.
N.P. Pavletich, B.S. Carver, C. Cordon-Cardo, H. Erdjument-Bromage, [236] X. Jiang, O. Charlat, R. Zamponi, Y. Yang, F. Cong, Dishevelled promotes Wnt
Ubiquitination regulates PTEN nuclear import and tumor suppression, Cell 128 receptor degradation through recruitment of ZNRF3/RNF43 E3 ubiquitin ligases,
(2007) 141–156. Mol. Cell 58 (2015) 522–533.
[211] X. Wang, X. Jiang, Post-translational regulation of PTEN, Oncogene 27 (2008) [237] A. Loregger, M. Grandl, R. Mejias-Luque, M. Allgauer, K. Degenhart,
5454–5463. V. Haselmann, C. Oikonomou, P. Hatzis, K.P. Janssen, U. Nitsche, D. Gradl, O. van
[212] X. Wang, L.C. Trotman, T. Koppie, A. Alimonti, Z. Chen, Z. Gao, J. Wang, den Broek, O. Destree, K. Ulm, M. Neumaier, B. Kalali, A. Jung, I. Varela,
H. Erdjument-Bromage, P. Tempst, C. Cordon-Cardo, NEDD4-1 is a proto-onco- R.M. Schmid, R. Rad, D.H. Busch, M. Gerhard, The E3 ligase RNF43 inhibits Wnt
genic ubiquitin ligase for PTEN, Cell 128 (2007) 129–139. signaling downstream of mutated beta-catenin by sequestering TCF4 to the nu-
[213] N. Amodio, M. Scrima, L. Palaia, A.N. Salman, A. Quintiero, R. Franco, G. Botti, clear membrane, Sci. Signal. 8 (2015) ra90.
P. Pirozzi, G. Rocco, N. De Rosa, G. Viglietto, Oncogenic role of the E3 ubiquitin [238] A. Mukai, M. Yamamoto-Hino, W. Awano, W. Watanabe, M. Komada, S. Goto,
ligase NEDD4-1, a PTEN negative regulator, in non-small-cell lung carcinomas, Balanced ubiquitylation and deubiquitylation of frizzled regulate cellular re-
Am. J. Pathol. 177 (2010) 2622–2634. sponsiveness to Wg/Wnt, EMBO J. 29 (2010) 2114–2125.
[214] F. Fouladkou, T. Landry, H. Kawabe, A. Neeb, C. Lu, N. Brose, V. Stambolic, [239] B. Madan, M.P. Walker, R. Young, L. Quick, K.A. Orgel, M. Ryan, P. Gupta,
D. Rotin, The ubiquitin ligase Nedd4-1 is dispensable for the regulation of PTEN I.C. Henrich, M. Ferrer, S. Marine, B.S. Roberts, W.T. Arthur, J.D. Berndt,
stability and localization, Proc. Natl. Acad. Sci. 105 (2008) 8585–8590. A.M. Oliveira, R.T. Moon, D.M. Virshup, M.M. Chou, M.B. Major, USP6 oncogene
[215] M.S. Song, L. Salmena, A. Carracedo, A. Egia, F. Lo-Coco, J. Teruya-Feldstein, promotes Wnt signaling by deubiquitylating Frizzleds, Proc. Natl. Acad. Sci. U. S.
P.P. Pandolfi, The deubiquitinylation and localization of PTEN are regulated by a A. 113 (2016) E2945–2954.
HAUSP–PML network, Nature 455 (2008) 813–817. [240] T. Schwarz-Romond, M. Fiedler, N. Shibata, P.J. Butler, A. Kikuchi, Y. Higuchi,
[216] C. Van Themsche, V. Leblanc, S. Parent, E. Asselin, X-linked inhibitor of apoptosis M. Bienz, The DIX domain of dishevelled confers Wnt signaling by dynamic
protein (XIAP) regulates PTEN ubiquitination, content, and compartmentalization, polymerization, Nat. Struct. Mol. Biol. 14 (2007) 484–492.
J. Biol. Chem. 284 (2009) 20462–20466. [241] Y. Zhang, S. Liu, C. Mickanin, Y. Feng, O. Charlat, G.A. Michaud, M. Schirle, X. Shi,
[217] S. Maddika, S. Kavela, N. Rani, V.R. Palicharla, J.L. Pokorny, J.N. Sarkaria, M. Hild, A. Bauer, V.E. Myer, P.M. Finan, J.A. Porter, S.M. Huang, F. Cong,
J. Chen, WWP2 is an E3 ubiquitin ligase for PTEN, Nat. Cell Biol. 13 (2011) RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation
728–733. and Wnt signalling, Nat. Cell Biol. 13 (2011) 623–629.
[218] S.F. Ahmed, S. Deb, I. Paul, A. Chatterjee, T. Mandal, U. Chatterjee, M.K. Ghosh, [242] S. Kim, E.H. Jho, The protein stability of Axin, a negative regulator of Wnt sig-
The chaperone-assisted E3 ligase C terminus of Hsc70-interacting protein (CHIP) naling, is regulated by Smad ubiquitination regulatory factor 2 (Smurf2), J. Biol.
targets PTEN for proteasomal degradation, J. Biol. Chem. 287 (2012) Chem. 285 (2010) 36420–36426.
15996–16006. [243] C. Fei, X. He, S. Xie, H. Miao, Z. Zhou, L. Li, Smurf1-mediated axin ubiquitination
[219] J.T. Lee, J. Shan, J. Zhong, M. Li, B. Zhou, A. Zhou, R. Parsons, W. Gu, RFP- requires Smurf1 C2 domain and is cell cycle-dependent, J. Biol. Chem. 289 (2014)
mediated ubiquitination of PTEN modulates its effect on AKT activation, Cell Res. 14170–14177.
23 (2013) 552–564. [244] T.T. Lui, C. Lacroix, S.M. Ahmed, S.J. Goldenberg, C.A. Leach, A.M. Daulat,
[220] J. Zhang, P. Zhang, Y. Wei, H.L. Piao, W. Wang, S. Maddika, M. Wang, D. Chen, S. Angers, The ubiquitin-specific protease USP34 regulates axin stability and Wnt/
Y. Sun, M.C. Hung, J. Chen, L. Ma, Deubiquitylation and stabilization of PTEN by beta-catenin signaling, Mol. Cell. Biol. 31 (2011) 2053–2065.
USP13, Nat. Cell Biol. 15 (2013) 1486–1494. [245] H. Tran, F. Hamada, T. Schwarz-Romond, M. Bienz, Trabid, a new positive reg-
[221] L. Yuan, Y. Lv, H. Li, H. Gao, S. Song, Y. Zhang, G. Xing, X. Kong, L. Wang, Y. Li, ulator of Wnt-induced transcription with preference for binding and cleaving K63-
T. Zhou, D. Gao, Z.X. Xiao, Y. Yin, W. Wei, F. He, L. Zhang, Deubiquitylase OTUD3 linked ubiquitin chains, Genes Dev. 22 (2008) 528–542.
regulates PTEN stability and suppresses tumorigenesis, Nat. Cell Biol. 17 (2015) [246] S.I. Yun, H.H. Kim, J.H. Yoon, W.S. Park, M.J. Hahn, H.C. Kim, C.H. Chung,
1169–1181. K.K. Kim, Ubiquitin specific protease 4 positively regulates the WNT/beta-catenin
[222] L. Yuan, Y. Lv, H. Li, H. Gao, S. Song, Y. Zhang, G. Xing, X. Kong, L. Wang, Y. Li, signaling in colorectal cancer, Mol. Oncol. 9 (2015) 1834–1851.
Deubiquitylase OTUD3 regulates PTEN stability and suppresses tumorigenesis, [247] B. Zhao, C. Schlesiger, M.G. Masucci, K. Lindsten, The ubiquitin specific protease 4
Nat. Cell Biol. (2015). (USP4) is a new player in the Wnt signalling pathway, J. Cell. Mol. Med. 13 (2009)
[223] O. Tetsu, F. McCormick, Beta-catenin regulates expression of cyclin D1 in colon 1886–1895.
carcinoma cells, Nature 398 (1999) 422–426. [248] J. Shi, Y. Liu, X. Xu, W. Zhang, T. Yu, J. Jia, C. Liu, Deubiquitinase USP47/UBP64E
[224] T.C. He, A.B. Sparks, C. Rago, H. Hermeking, L. Zawel, L.T. da Costa, P.J. Morin, regulates beta-catenin ubiquitination and degradation and plays a positive role in
B. Vogelstein, K.W. Kinzler, Identification of c-MYC as a target of the APC Wnt signaling, Mol. Cell. Biol. 35 (2015) 3301–3311.
pathway, Science 281 (1998) 1509–1512. [249] M.B. Greenblatt, D.Y. Shin, H. Oh, K.Y. Lee, B. Zhai, S.P. Gygi, S. Lotinun,
[225] D. Kimelman, W. Xu, Beta-catenin destruction complex: insights and questions R. Baron, D. Liu, B. Su, L.H. Glimcher, J.H. Shim, MEKK2 mediates an alternative
from a structural perspective, Oncogene 25 (2006) 7482–7491. beta-catenin pathway that promotes bone formation, Proc. Natl. Acad. Sci. U. S. A.
[226] M. Hart, J.P. Concordet, I. Lassot, I. Albert, R. del los Santos, H. Durand, C. Perret, 113 (2016) E1226–1235.
B. Rubinfeld, F. Margottin, R. Benarous, P. Polakis, The F-box protein beta-TrCP [250] P. Ma, X. Yang, Q. Kong, C. Li, S. Yang, Y. Li, B. Mao, The ubiquitin ligase RNF220
associates with phosphorylated beta-catenin and regulates its activity in the cell, enhances canonical Wnt signaling through USP7-mediated deubiquitination of
Curr. Biol. 9 (1999) 207–210. beta-catenin, Mol. Cell. Biol. 34 (2014) 4355–4366.
[227] V. Chitalia, S. Shivanna, J. Martorell, R. Meyer, E. Edelman, N. Rahimi, c-Cbl, a [251] P. D'Arcy, X. Wang, S. Linder, Deubiquitinase inhibition as a cancer therapeutic
ubiquitin E3 ligase that targets active beta-catenin: a novel layer of Wnt signaling strategy, Pharmacol. Ther. 147 (2015) 32–54.
regulation, J. Biol. Chem. 288 (2013) 23505–23517. [252] E. Koulich, X. Li, G.N. DeMartino, Relative structural and functional roles of
[228] S. Shivanna, I. Harrold, M. Shashar, R. Meyer, C. Kiang, J. Francis, Q. Zhao, multiple deubiquitylating proteins associated with mammalian 26S proteasome,
H. Feng, E.R. Edelman, N. Rahimi, V.C. Chitalia, The c-Cbl ubiquitin ligase reg- Mol. Biol. Cell 19 (2008) 1072–1082.
ulates nuclear beta-catenin and angiogenesis by its tyrosine phosphorylation [253] B.H. Lee, Y. Lu, M.A. Prado, Y. Shi, G. Tian, S. Sun, S. Elsasser, S.P. Gygi,
mediated through the Wnt signaling pathway, J. Biol. Chem. 290 (2015) R.W. King, D. Finley, USP14 deubiquitinates proteasome-bound substrates that are
12537–12546. ubiquitinated at multiple sites, Nature 532 (2016) 398–401.
[229] V.C. Chitalia, R.L. Foy, M.M. Bachschmid, L. Zeng, M.V. Panchenko, M.I. Zhou, [254] B.H. Lee, M.J. Lee, S. Park, D.C. Oh, S. Elsasser, P.C. Chen, C. Gartner, N. Dimova,
A. Bharti, D.C. Seldin, S.H. Lecker, I. Dominguez, H.T. Cohen, Jade-1 inhibits Wnt J. Hanna, S.P. Gygi, S.M. Wilson, R.W. King, D. Finley, Enhancement of protea-
signalling by ubiquitylating beta-catenin and mediates Wnt pathway inhibition by some activity by a small-molecule inhibitor of USP14, Nature 467 (2010)
pVHL, Nat. Cell Biol. 10 (2008) 1208–1216. 179–184.
[230] R.E. de Groot, R.S. Ganji, O. Bernatik, B. Lloyd-Lewis, K. Seipel, K. Sedova, [255] P.C. Chen, L.N. Qin, X.M. Li, B.J. Walters, J.A. Wilson, L. Mei, S.M. Wilson, The
Z. Zdrahal, V.M. Dhople, T.C. Dale, H.C. Korswagen, V. Bryja, Huwe1-mediated proteasome-associated deubiquitinating enzyme Usp14 is essential for the main-
ubiquitylation of dishevelled defines a negative feedback loop in the Wnt signaling tenance of synaptic ubiquitin levels and the development of neuromuscular
pathway, Sci. Signal. 7 (2014) ra26. junctions, The Journal of Neuroscience: The Official Journal of the Society for

481
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

Neuroscience 29 (2009) 10909–10919. identification of an oxidation-sensitive subunit in 26 S proteasome, Biochemistry


[256] S.M. Wilson, B. Bhattacharyya, R.A. Rachel, V. Coppola, L. Tessarollo, 44 (2005) 13893–13901.
D.B. Householder, C.F. Fletcher, R.J. Miller, N.G. Copeland, N.A. Jenkins, Synaptic [282] H. Liu, W. Li, M. Ahmad, T.M. Miller, M.E. Rose, S.M. Poloyac, G. Uechi,
defects in ataxia mice result from a mutation in Usp14, encoding a ubiquitin- M. Balasubramani, R.W. Hickey, S.H. Graham, Modification of ubiquitin-C-term-
specific protease, Nat. Genet. 32 (2002) 420–425. inal hydrolase-L1 by cyclopentenone prostaglandins exacerbates hypoxic injury,
[257] S. Hussain, T. Bedekovics, M. Chesi, P.L. Bergsagel, P.J. Galardy, UCHL1 is a Neurobiol. Dis. 41 (2011) 318–328.
biomarker of aggressive multiple myeloma required for disease progression, [283] Z. Li, F. Melandri, I. Berdo, M. Jansen, L. Hunter, S. Wright, D. Valbrun,
Oncotarget 6 (2015) 40704–40718. M.E. Figueiredo-Pereira, Delta12-prostaglandin J2 inhibits the ubiquitin hydrolase
[258] Y. Goto, L. Zeng, C.J. Yeom, Y. Zhu, A. Morinibu, K. Shinomiya, M. Kobayashi, UCH-L1 and elicits ubiquitin-protein aggregation without proteasome inhibition,
K. Hirota, S. Itasaka, M. Yoshimura, K. Tanimoto, M. Torii, T. Sowa, T. Menju, Biochem. Biophys. Res. Commun. 319 (2004) 1171–1180.
M. Sonobe, H. Kakeya, M. Toi, H. Date, E.M. Hammond, M. Hiraoka, H. Harada, [284] L.M. Koharudin, H. Liu, R. Di Maio, R.B. Kodali, S.H. Graham, A.M. Gronenborn,
UCHL1 provides diagnostic and antimetastatic strategies due to its deubiquiti- Cyclopentenone prostaglandin-induced unfolding and aggregation of the
nating effect on HIF-1alpha, Nat. Commun. 6 (2015) 6153. Parkinson disease-associated UCH-L1, Proc. Natl. Acad. Sci. U. S. A. 107 (2010)
[259] Y. Liu, H.A. Lashuel, S. Choi, X. Xing, A. Case, J. Ni, L.A. Yeh, G.D. Cuny, 6835–6840.
R.L. Stein, P.T. Lansbury Jr., Discovery of inhibitors that elucidate the role of UCH- [285] H. Erdal, M. Berndtsson, J. Castro, U. Brunk, M.C. Shoshan, S. Linder, Induction of
L1 activity in the H1299 lung cancer cell line, Chem. Biol. 10 (2003) 837–846. lysosomal membrane permeabilization by compounds that activate p53-in-
[260] Y.Y. Tan, H.Y. Zhou, Z.Q. Wang, S.D. Chen, Endoplasmic reticulum stress con- dependent apoptosis, Proc. Natl. Acad. Sci. U. S. A. 102 (2005) 192–197.
tributes to the cell death induced by UCH-L1 inhibitor, Mol. Cell. Biochem. 318 [286] M. Berndtsson, M. Beaujouin, L. Rickardson, A.M. Havelka, R. Larsson,
(2008) 109–115. J. Westman, E. Liaudet-Coopman, S. Linder, Induction of the lysosomal apoptosis
[261] A.H. Mermerian, A. Case, R.L. Stein, G.D. Cuny, Structure-activity relationship, pathway by inhibitors of the ubiquitin-proteasome system, Int. J. Cancer 124
kinetic mechanism, and selectivity for a new class of ubiquitin C-terminal hy- (2009) 1463–1469.
drolase-L1 (UCH-L1) inhibitors, Bioorg. Med. Chem. Lett. 17 (2007) 3729–3732. [287] Z. Tian, P. D'Arcy, X. Wang, A. Ray, Y.T. Tai, Y. Hu, R.D. Carrasco, P. Richardson,
[262] J. Sosna, S. Philipp, J. Fuchslocher Chico, C. Saggau, J. Fritsch, A. Foll, J. Plenge, S. Linder, D. Chauhan, K.C. Anderson, A novel small molecule inhibitor of deu-
C. Arenz, T. Pinkert, H. Kalthoff, A. Trauzold, I. Schmitz, S. Schutze, D. Adam, biquitylating enzyme USP14 and UCHL5 induces apoptosis in multiple myeloma
Differences and similarities in TRAIL- and tumor necrosis factor-mediated ne- and overcomes bortezomib resistance, Blood 123 (2014) 706–716.
croptotic signaling in cancer cells, Mol. Cell. Biol. 36 (2016) 2626–2644. [288] X. Wang, M. Mazurkiewicz, E.K. Hillert, M.H. Olofsson, S. Pierrou, P. Hillertz,
[263] X. Tian, N.S. Isamiddinova, R.J. Peroutka, S.J. Goldenberg, M.R. Mattern, J. Gullbo, K. Selvaraju, A. Paulus, S. Akhtar, F. Bossler, A.C. Khan, S. Linder,
B. Nicholson, C. Leach, Characterization of selective ubiquitin and ubiquitin-like P. D'Arcy, The proteasome deubiquitinase inhibitor VLX1570 shows selectivity for
protease inhibitors using a fluorescence-based multiplex assay format, Assay Drug ubiquitin-specific protease-14 and induces apoptosis of multiple myeloma cells,
Dev. Technol. 9 (2011) 165–173. Sci Rep 6 (2016) 26979.
[264] J. Weinstock, J. Wu, P. Cao, W.D. Kingsbury, J.L. McDermott, M.P. Kodrasov, [289] P. D'Arcy, S. Brnjic, M.H. Olofsson, M. Fryknas, K. Lindsten, M. De Cesare,
D.M. McKelvey, K.G. Suresh Kumar, S.J. Goldenberg, M.R. Mattern, B. Nicholson, P. Perego, B. Sadeghi, M. Hassan, R. Larsson, S. Linder, Inhibition of proteasome
Selective dual inhibitors of the cancer-related deubiquitylating proteases USP7 deubiquitinating activity as a new cancer therapy, Nat. Med. 17 (2011)
and USP47, ACS Med. Chem. Lett. 3 (2012) 789–792. 1636–1640.
[265] C. Reverdy, S. Conrath, R. Lopez, C. Planquette, C. Atmanene, V. Collura, [290] E. Aleo, C.J. Henderson, A. Fontanini, B. Solazzo, C. Brancolini, Identification of
J. Harpon, V. Battaglia, V. Vivat, W. Sippl, F. Colland, Discovery of specific in- new compounds that trigger apoptosome-independent caspase activation and
hibitors of human USP7/HAUSP deubiquitinating enzyme, Chem. Biol. 19 (2012) apoptosis, Cancer Res. 66 (2006) 9235–9244.
467–477. [291] A. Fontanini, C. Foti, H. Potu, E. Crivellato, R. Maestro, P. Bernardi, F. Demarchi,
[266] S.M. Nijman, T.T. Huang, A.M. Dirac, T.R. Brummelkamp, R.M. Kerkhoven, C. Brancolini, The isopeptidase inhibitor G5 triggers a caspase-independent ne-
A.D. D'Andrea, R. Bernards, The deubiquitinating enzyme USP1 regulates the crotic death in cells resistant to apoptosis: a comparative study with the protea-
Fanconi anemia pathway, Mol. Cell 17 (2005) 331–339. some inhibitor bortezomib, J. Biol. Chem. 284 (2009) 8369–8381.
[267] K. Yang, G.L. Moldovan, P. Vinciguerra, J. Murai, S. Takeda, A.D. D'Andrea, [292] B. Nicholson, C.A. Leach, S.J. Goldenberg, D.M. Francis, M.P. Kodrasov, X. Tian,
Regulation of the Fanconi anemia pathway by a SUMO-like delivery network, J. Shanks, D.E. Sterner, A. Bernal, M.R. Mattern, K.D. Wilkinson, T.R. Butt,
Genes Dev. 25 (2011) 1847–1858. Characterization of ubiquitin and ubiquitin-like-protein isopeptidase activities,
[268] J. Chen, T.S. Dexheimer, Y. Ai, Q. Liang, M.A. Villamil, J. Inglese, D.J. Maloney, Protein Sci. 17 (2008) 1035–1043.
A. Jadhav, A. Simeonov, Z. Zhuang, Selective and cell-active inhibitors of the [293] R.K. Anchoori, S.R. Khan, T. Sueblinvong, A. Felthauser, Y. Iizuka, R. Gavioli,
USP1/UAF1 deubiquitinase complex reverse cisplatin resistance in non-small cell F. Destro, R. Isaksson Vogel, S. Peng, R.B. Roden, M. Bazzaro, Stressing the ubi-
lung cancer cells, Chem. Biol. 18 (2011) 1390–1400. quitin-proteasome system without 20S proteolytic inhibition selectively kills cer-
[269] S. Ohayon, L. Spasser, A. Aharoni, A. Brik, Targeting deubiquitinases enabled by vical cancer cells, PLoS One 6 (2011) e23888.
chemical synthesis of proteins, J. Am. Chem. Soc. 134 (2012) 3281–3289. [294] O.A. Issaenko, A.Y. Amerik, Chalcone-based small-molecule inhibitors attenuate
[270] V. Seiberlich, O. Goldbaum, V. Zhukareva, C. Richter-Landsberg, The small mo- malignant phenotype via targeting deubiquitinating enzymes, Cell Cycle 11
lecule inhibitor PR-619 of deubiquitinating enzymes affects the microtubule net- (2012) 1804–1817.
work and causes protein aggregate formation in neural cells: implications for [295] H.Z. Zhang, S. Kasibhatla, Y. Wang, J. Herich, J. Guastella, B. Tseng, J. Drewe,
neurodegenerative diseases, Biochim. Biophys. Acta 1823 (2012) 2057–2068. S.X. Cai, Discovery, characterization and SAR of gambogic acid as a potent
[271] V. Seiberlich, J. Borchert, V. Zhukareva, C. Richter-Landsberg, Inhibition of pro- apoptosis inducer by a HTS assay, Bioorg. Med. Chem. 12 (2004) 309–317.
tein deubiquitination by PR-619 activates the autophagic pathway in OLN-t40 [296] J. Felth, K. Lesiak-Mieczkowska, P. D'Arcy, C. Haglund, J. Gullbo, R. Larsson,
oligodendroglial cells, Cell Biochem. Biophys. 67 (2013) 149–160. S. Linder, L. Bohlin, M. Fryknas, L. Rickardson, Gambogic acid is cytotoxic to
[272] V. Kapuria, L.F. Peterson, D. Fang, W.G. Bornmann, M. Talpaz, N.J. Donato, cancer cells through inhibition of the ubiquitin-proteasome system, Investig. New
Deubiquitinase inhibition by small-molecule WP1130 triggers aggresome forma- Drugs 31 (2013) 587–598.
tion and tumor cell apoptosis, Cancer Res. 70 (2010) 9265–9276. [297] E. Pisha, H. Chai, I.S. Lee, T.E. Chagwedera, N.R. Farnsworth, G.A. Cordell,
[273] L.F. Peterson, H. Sun, Y. Liu, H. Potu, M. Kandarpa, M. Ermann, S.M. Courtney, C.W. Beecher, H.H. Fong, A.D. Kinghorn, D.M. Brown, et al., Discovery of betulinic
M. Young, H.D. Showalter, D. Sun, A. Jakubowiak, S.N. Malek, M. Talpaz, acid as a selective inhibitor of human melanoma that functions by induction of
N.J. Donato, Targeting deubiquitinase activity with a novel small-molecule in- apoptosis, Nat. Med. 1 (1995) 1046–1051.
hibitor as therapy for B-cell malignancies, Blood 125 (2015) 3588–3597. [298] F.B. Mullauer, J.H. Kessler, J.P. Medema, Betulinic acid, a natural compound with
[274] J.W. Perry, M. Ahmed, K.O. Chang, N.J. Donato, H.D. Showalter, C.E. Wobus, potent anticancer effects, Anti-Cancer Drugs 21 (2010) 215–227.
Antiviral activity of a small molecule deubiquitinase inhibitor occurs via induction [299] T. Reiner, R. Parrondo, A. de Las Pozas, D. Palenzuela, C. Perez-Stable, Betulinic
of the unfolded protein response, PLoS Pathog. 8 (2012) e1002783. acid selectively increases protein degradation and enhances prostate cancer-spe-
[275] L.V. Pham, A.T. Tamayo, C. Li, W. Bornmann, W. Priebe, R.J. Ford, Degrasyn cific apoptosis: possible role for inhibition of deubiquitinase activity, PLoS One 8
potentiates the antitumor effects of bortezomib in mantle cell lymphoma cells in (2013) e56234.
vitro and in vivo: therapeutic implications, Mol. Cancer Ther. 9 (2010) [300] S. D'Aguanno, I. D'Agnano, M. De Canio, C. Rossi, S. Bernardini, G. Federici,
2026–2036. A. Urbani, Shotgun proteomics and network analysis of neuroblastoma cell lines
[276] E. Fiebiger, C. Hirsch, J.M. Vyas, E. Gordon, H.L. Ploegh, D. Tortorella, Dissection treated with curcumin, Mol. BioSyst. 8 (2012) 1068–1077.
of the dislocation pathway for type I membrane proteins with a new small mo- [301] B. Zhou, Y. Zuo, B. Li, H. Wang, H. Liu, X. Wang, X. Qiu, Y. Hu, S. Wen, J. Du,
lecule inhibitor, eeyarestatin, Mol. Biol. Cell 15 (2004) 1635–1646. X. Bu, Deubiquitinase inhibition of 19S regulatory particles by 4-arylidene cur-
[277] Q. Wang, L. Li, Y. Ye, Inhibition of p97-dependent protein degradation by cumin analog AC17 causes NF-kappaB inhibition and p53 reactivation in human
eeyarestatin I, J. Biol. Chem. 283 (2008) 7445–7454. lung cancer cells, Mol. Cancer Ther. 12 (2013) 1381–1392.
[278] J.E. Mullally, P.J. Moos, K. Edes, F.A. Fitzpatrick, Cyclopentenone prostaglandins [302] A. Ernst, G. Avvakumov, J. Tong, Y. Fan, Y. Zhao, P. Alberts, A. Persaud,
of the J series inhibit the ubiquitin isopeptidase activity of the proteasome J.R. Walker, A.M. Neculai, D. Neculai, A. Vorobyov, P. Garg, L. Beatty, P.K. Chan,
pathway, J. Biol. Chem. 276 (2001) 30366–30373. Y.C. Juang, M.C. Landry, C. Yeh, E. Zeqiraj, K. Karamboulas, A. Allali-Hassani,
[279] F.A. Fitzpatrick, M.A. Wynalda, Albumin-catalyzed metabolism of prostaglandin M. Vedadi, M. Tyers, J. Moffat, F. Sicheri, L. Pelletier, D. Durocher, B. Raught,
D2. Identification of products formed in vitro, J. Biol. Chem. 258 (1983) D. Rotin, J. Yang, M.F. Moran, S. Dhe-Paganon, S.S. Sidhu, A strategy for mod-
11713–11718. ulation of enzymes in the ubiquitin system, Science 339 (2013) 590–595.
[280] J.E. Mullally, F.A. Fitzpatrick, Pharmacophore model for novel inhibitors of ubi- [303] W. Zhang, M.A. Sartori, T. Makhnevych, K.E. Federowicz, X. Dong, L. Liu, S. Nim,
quitin isopeptidases that induce p53-independent cell death, Mol. Pharmacol. 62 A. Dong, J. Yang, Y. Li, D. Haddad, A. Ernst, D. Heerding, Y. Tong, J. Moffat,
(2002) 351–358. S.S. Sidhu, Generation and validation of intracellular ubiquitin variant inhibitors
[281] T. Ishii, T. Sakurai, H. Usami, K. Uchida, Oxidative modification of proteasome: for USP7 and USP10, J. Mol. Biol. (2017).

482
N. Kumari et al. BBA - Reviews on Cancer 1868 (2017) 456–483

[304] N. Cancer Genome Atlas Research, Comprehensive molecular characterization of Systems, C. Memorial Sloan Kettering Cancer, C. University of California Santa,
urothelial bladder carcinoma, Nature 507 (2014) 315–322. M.D.A.C.C. University of Texas, C. Biospecimen Core Resource: International
[305] B. Pereira, S.F. Chin, O.M. Rueda, H.K. Vollan, E. Provenzano, H.A. Bardwell, Genomics, H. Research Institute at Nationwide Children's, S. Tissue Source Sites:
M. Pugh, L. Jones, R. Russell, S.J. Sammut, D.W. Tsui, B. Liu, S.J. Dawson, Analytic Biologic, C. Asan Medical, B. Asterand, H. Barretos Cancer,
J. Abraham, H. Northen, J.F. Peden, A. Mukherjee, G. Turashvili, A.R. Green, BioreclamationIvt, C. Botkin Municipal, S. Chonnam National University Medical,
S. McKinney, A. Oloumi, S. Shah, N. Rosenfeld, L. Murphy, D.R. Bentley, I.O. Ellis, S. Christiana Care Health, Cureline, U. Duke, U. Emory, U. Erasmus, M. Indiana
A. Purushotham, S.E. Pinder, A.L. Borresen-Dale, H.M. Earl, P.D. Pharoah, University School of, M. Institute of Oncology of, C. International Genomics,
M.T. Ross, S. Aparicio, C. Caldas, The somatic mutation profiles of 2,433 breast Invidumed, H. Israelitisches Krankenhaus, M. Keimyung University School of, C.
cancers refines their genomic and transcriptomic landscapes, Nat. Commun. 7 Memorial Sloan Kettering Cancer, G. National Cancer Center, B. Ontario Tumour,
(2016) 11479. C. Peter MacCallum Cancer, S. Pusan National University Medical, S. Ribeirao
[306] N. Cancer Genome Atlas, Comprehensive molecular portraits of human breast Preto Medical, H. St. Joseph's, C. Medical, U. St. Petersburg Academic, B. Tayside
tumours, Nature 490 (2012) 61–70. Tissue, D. University of, C. University of Kansas Medical, M. University of, H.
[307] M. Ceccarelli, F.P. Barthel, T.M. Malta, T.S. Sabedot, S.R. Salama, B.A. Murray, University of North Carolina at Chapel, M. University of Pittsburgh School of,
O. Morozova, Y. Newton, A. Radenbaugh, S.M. Pagnotta, S. Anjum, J. Wang, M.D.A.C.C. University of Texas, U. Disease Working Group: Duke, C. Memorial
G. Manyam, P. Zoppoli, S. Ling, A.A. Rao, M. Grifford, A.D. Cherniack, H. Zhang, Sloan Kettering Cancer, I. National Cancer, M.D.A.C.C. University of Texas, M.
L. Poisson, C.G. Carlotti Jr., D.P. Tirapelli, A. Rao, T. Mikkelsen, C.C. Lau, Yonsei University College of, C.I. Data Coordination Center, H. Project Team:
W.K. Yung, R. Rabadan, J. Huse, D.J. Brat, N.L. Lehman, J.S. Barnholtz-Sloan, National Institutes of, Integrated genomic characterization of oesophageal carci-
S. Zheng, K. Hess, G. Rao, M. Meyerson, R. Beroukhim, L. Cooper, R. Akbani, noma, Nature 541 (2017) 169–175.
M. Wrensch, D. Haussler, K.D. Aldape, P.W. Laird, D.H. Gutmann, T.R. Network, [310] N. Cancer Genome Atlas Research, Comprehensive molecular characterization of
H. Noushmehr, A. Iavarone, R.G. Verhaak, Molecular profiling reveals biologically gastric adenocarcinoma, Nature 513 (2014) 202–209.
discrete subsets and pathways of progression in diffuse glioma, Cell 164 (2016) [311] N. Cancer Genome Atlas Research, Comprehensive molecular characterization of
550–563. clear cell renal cell carcinoma, Nature 499 (2013) 43–49.
[308] C.W. Brennan, R.G. Verhaak, A. McKenna, B. Campos, H. Noushmehr, S.R. Salama, [312] N. Cancer Genome Atlas Research, Comprehensive molecular profiling of lung
S. Zheng, D. Chakravarty, J.Z. Sanborn, S.H. Berman, R. Beroukhim, B. Bernard, adenocarcinoma, Nature 511 (2014) 543–550.
C.J. Wu, G. Genovese, I. Shmulevich, J. Barnholtz-Sloan, L. Zou, R. Vegesna, [313] N. Cancer Genome Atlas Research, The molecular taxonomy of primary prostate
S.A. Shukla, G. Ciriello, W.K. Yung, W. Zhang, C. Sougnez, T. Mikkelsen, cancer, Cell 163 (2015) 1011–1025.
K. Aldape, D.D. Bigner, E.G. Van Meir, M. Prados, A. Sloan, K.L. Black, [314] J. Barretina, B.S. Taylor, S. Banerji, A.H. Ramos, M. Lagos-Quintana,
J. Eschbacher, G. Finocchiaro, W. Friedman, D.W. Andrews, A. Guha, M. Iacocca, P.L. Decarolis, K. Shah, N.D. Socci, B.A. Weir, A. Ho, D.Y. Chiang, B. Reva,
B.P. O'Neill, G. Foltz, J. Myers, D.J. Weisenberger, R. Penny, R. Kucherlapati, C.H. Mermel, G. Getz, Y. Antipin, R. Beroukhim, J.E. Major, C. Hatton, R. Nicoletti,
C.M. Perou, D.N. Hayes, R. Gibbs, M. Marra, G.B. Mills, E. Lander, P. Spellman, M. Hanna, T. Sharpe, T.J. Fennell, K. Cibulskis, R.C. Onofrio, T. Saito, N. Shukla,
R. Wilson, C. Sander, J. Weinstein, M. Meyerson, S. Gabriel, P.W. Laird, C. Lau, S. Nelander, S.J. Silver, C. Sougnez, A. Viale, W. Winckler, R.G. Maki,
D. Haussler, G. Getz, L. Chin, T.R. Network, The somatic genomic landscape of L.A. Garraway, A. Lash, H. Greulich, D.E. Root, W.R. Sellers, G.K. Schwartz,
glioblastoma, Cell 155 (2013) 462–477. C.R. Antonescu, E.S. Lander, H.E. Varmus, M. Ladanyi, C. Sander, M. Meyerson,
[309] N. Cancer Genome Atlas Research, U. Analysis Working Group, Asan, B.C.C. S. Singer, Subtype-specific genomic alterations define new targets for soft-tissue
Agency, Brigham, H. Women's, I. Broad, U. Brown, U. Case Western Reserve, I. sarcoma therapy, Nat. Genet. 42 (2010) 715–721.
Dana-Farber Cancer, U. Duke, C. Greater Poland Cancer, S. Harvard Medical, B. [315] N. Cancer Genome Atlas, Comprehensive genomic characterization of head and
Institute for Systems, K.U. Leuven, C. Mayo, C. Memorial Sloan Kettering Cancer, I. neck squamous cell carcinomas, Nature 517 (2015) 576–582.
National Cancer, H. Nationwide Children's, U. Stanford, A. University of, M. [316] A.K. Witkiewicz, E.A. McMillan, U. Balaji, G. Baek, W.C. Lin, J. Mansour,
University of, C. University of North, P. University of, R. University of, C. M. Mollaee, K.U. Wagner, P. Koduru, A. Yopp, M.A. Choti, C.J. Yeo, P. McCue,
University of Southern, M.D.A.C.C. University of Texas, W. University of, I. Van M.A. White, E.S. Knudsen, Whole-exome sequencing of pancreatic cancer defines
Andel Research, U. Vanderbilt, U. Washington, I. Genome Sequencing Center: genetic diversity and therapeutic targets, Nat. Commun. 6 (2015) 6744.
Broad, L. Washington University in St, B.C.C.A. Genome Characterization Centers, [317] N. Cancer Genome Atlas Research, C. Kandoth, N. Schultz, A.D. Cherniack,
I. Broad, S. Harvard Medical, U. Sidney Kimmel Comprehensive Cancer Center at R. Akbani, Y. Liu, H. Shen, A.G. Robertson, I. Pashtan, R. Shen, C.C. Benz, C. Yau,
Johns Hopkins, C. University of North, C. University of Southern California P.W. Laird, L. Ding, W. Zhang, G.B. Mills, R. Kucherlapati, E.R. Mardis,
Epigenome, M.D.A.C.C. University of Texas, I. Van Andel Research, I. Genome D.A. Levine, Integrated genomic characterization of endometrial carcinoma,
Data Analysis Centers: Broad, U. Brown, S. Harvard Medical, B. Institute for Nature 497 (2013) 67–73.

483

Potrebbero piacerti anche