You are on page 1of 11

The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337

Contents lists available at SciVerse ScienceDirect

The International Journal of Biochemistry


& Cell Biology
journal homepage: www.elsevier.com/locate/biocel

Review

Deregulation of the COP9 signalosome–cullin-RING ubiquitin-ligase


pathway: Mechanisms and roles in urological cancers
Linda Gummlich a,b , Anja Rabien c , Klaus Jung b,c , Wolfgang Dubiel a,∗
a
Department of General, Visceral, Vascular and Thoracic Surgery, Division of Molecular Biology, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117
Berlin, Germany
b
Berlin Institute for Urological Research, Berlin, Germany
c
Department of Urology, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany

a r t i c l e i n f o a b s t r a c t

Article history: The COP9 signalosome (CSN)–cullin-RING ubiquitin (Ub)-ligase (CRL) pathway is a prominent segment
Received 30 January 2013 of the Ub proteasome system (UPS). It specifically ubiquitinates proteins and targets them for proteolytic
Received in revised form 21 March 2013 elimination. As part of the UPS it maintains essential cellular processes including cell cycle progression,
Accepted 22 March 2013
DNA repair, antigen processing and signal transduction. The CSN–CRL pathway consists of the CSN pos-
Available online 10 April 2013
sessing eight subunits (CSN1-CSN8) and one CRL consisting of a cullin, a RING-domain protein and a
substrate recognition subunit (SRS). In human cells approximately 250 CRLs exist each of which inter-
Keywords:
acting with a specific set of substrates and the CSN. The CSN–CRL interplay determines the activity and
COP9 signalosome
Cullin-RING ubiquitin ligases
specificity of CRL ubiquitination. The removal of the Ub-like protein Nedd8 from the CRL component cullin
Skp2 by the CSN (deneddylation) reduces the ubiquitinating activity and at the same time enables reassembly
Urological cancer of CRLs in order to adapt to substrate specificity requirements. On the other hand, CRLs as well as sub-
microRNAs strates negatively influence the deneddylating activity of the CSN. In recent years evidence accumulated
Tumor therapy that deregulation of the CSN–CRL pathway can cause cancer. Here we review current knowledge on mod-
ifications of CSN and CRL components including CSN subunits, SRSs and cullins causing tumorigenesis
with emphasis on urological neoplasia. The CSN–CRL pathway is a target of tumor-viruses as well as of
a multitude of miRNAs. Recently evaluated miRNAs altered in urological cancers might have impact on
the CSN–CRL pathway which has to be analyzed in future experiments. We propose that the pathway is
a suitable target for future tumor therapy.
© 2013 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1328
2. Architecture of CSN–CRL supercomplexes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1328
3. Regulation of the CSN–CRL pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1328
4. Deregulation of the CSN–CRL pathway and its role in tumorigenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1329
4.1. Deregulation of substrate recognition subunits . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1329
4.2. Deregulation of cullins, CAND1 and CSN subunits . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1330
4.3. Impact of tumor-viruses on the CSN–CRL pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1331
5. miRNA-mediated regulation of the CSN–CRL pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1332
5.1. Impact of the miRNome on the CSN–CRL complex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1332
5.2. Evaluated miRNAs altered in urological cancers and their possible impact on components of the CSN–CRL pathway . . . . . . . . . . . . . . . . . . . . 1333

Abbreviations: ␤-TrCP, ␤-transducin repeat-containing protein; CAND1, cullin-associated NEDD8-dissociated protein 1; CSN, COP9 signalosome; CRL, cullin-RING ubi-
quitin ligase; CUL, cullin; FBP, F-box protein; Fbw7, F-box protein WD40 domain 7; Keap1, Kelch-like ECH-associated protein 1; Nedd8, neural precursor cell expressed
developmentally down-regulated 8; miR, microRNA; mTOR, mammalian target of rapamycin; MPN, Mpr1p and Pad1p N-terminal; PCI, proteasome, COP9, and Initiation
factor 3; Rbx1, RING-box protein 1; RCC, renal cell carcinoma; Skp2, S-phase kinase-associated protein 2; SRS, substrate recognition subunit; Ub, ubiquitin; UPS, ubiquitin
proteasome system; VHL, von Hippel–Lindau protein.
∗ Corresponding author. Tel.: +49 30 450522305; fax: +49 30 450522928.
E-mail address: wolfgang.dubiel@charite.de (W. Dubiel).

1357-2725/$ – see front matter © 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.biocel.2013.03.023
1328 L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337

6. Is the CSN–CRL pathway a suitable target for cancer therapy? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1333


6.1. The deregulated CSN–CRL pathway as a target in cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1333
6.2. The CSN–CRL pathway component Skp2 might be a specific therapeutic target in urological cancers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1333
7. Future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1334
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1335
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1335

1. Introduction human CSN complex were performed by electron microscopy (EM)


(Kapelari et al., 2000). Images of the CSN and the lid were compared
In eukaryotic cells the dynamic state of intracellular proteins is revealing similar architectures of the two complexes. Integrating
mostly controlled by the ubiquitin (Ub) proteasome system (UPS). the EM data with known subunit–subunit interactions yielded the
In this system polyUb chains serve as markers for proteolysis. Ub first architectural model of the CSN (Kapelari et al., 2000). It has
is first activated by the Ub activating enzyme (E1). In a second been shown that the complex contains a single copy of each sub-
step activated Ub is transferred by the family of Ub conjugating unit and is composed of two symmetrical clusters, CSN1/2/3/8 and
enzymes (E2s) to a substrate supported by E3 Ub ligases. Selectivity CSN4/5/6/7, which are connected by CSN1-CSN6 (Sharon et al.,
and substrate specificity of the UPS is conferred by E3s specifically 2009). CRL1 complexes were crystallized (Goldenberg et al., 2004;
targeting proteins for degradation via ubiquitination (Hershko and Zheng et al., 2002) and molecular models of CRL1 action and
Ciechanover, 1998). The largest family of E3s are the cullin-RING CSN–CRL interaction were described (Deshaies and Joazeiro, 2009).
Ub ligases (CRLs) including approximately 250 members (Lee et al., The formation of CSN–CRL supercomplexes (Fig. 1B) has been
2011a). CRLs are multisubunit complexes composed of a cullin demonstrated in many organisms (Huang et al., 2009; Lyapina
from the CUL family (CUL-1-7), a RING domain subunit, mostly et al., 2001; Peng et al., 2003; Schwechheimer et al., 2001). Addi-
Rbx1, an adaptor protein and a substrate recognition subunit (SRS) tional progress in the field was recently obtained by EM and single
(Fig. 1A) (Petroski and Deshaies, 2005). In case of CUL-1 the SRSs particle analysis which generated a three-dimensional molecular
are F-box proteins (FBPs) and the complexes are called CRL1FBP . model for the CSN–CRL complex (Enchev et al., 2010, 2012). In
The activity of CRLs is regulated by conjugating and deconjugating this model, the PCI subunits of the CSN form an approximately
the Ub-like protein Nedd8 to cullins (Saha and Deshaies, 2008) (see coplanar surface. The four longest subunits (CSN1, CSN2, CSN3,
Chapter 3). Nedd8 activation and conjugation to proteins (cullins) and CSN4) are docked into the central region of an arc, which
occurs in a similar manner as described above for Ub (Liakopoulos is capped at each end by the shorter CSN7 and CSN8 subunits
et al., 1998). The COP9 signalosome (CSN) controls CRLs by specif- (Fig. 1A). The MPN domain subunits, CSN5 and CSN6, are pre-
ically deconjugating Nedd8 (deneddylation) from cullins (Fig. 1B) dicted to form a protrusion on the side opposite to the PCI subunits
(Bondar et al., 2006; Deshaies and Joazeiro, 2009; Lyapina et al., (Enchev et al., 2010, 2012; Sharon et al., 2009). In their model of
2001; Schwechheimer et al., 2001). The dynamic interplay and the CSN–CRL complex the authors show that the CSN predom-
activity of the CRLs and the CSN is called here the CSN–CRL path- inantly binds to the C-terminal portion of CUL-1 (Enchev et al.,
way and represents a pivotal element of the system that governs 2012). Neddylated CRL1 revealed a tighter binding to the CSN as
intracellular protein dynamics in eukaryotic cells. compared with the unneddylated one. As predicted before (Lyapina
The CSN is a conserved protein complex containing eight core et al., 2001), the molecular model of Enchev et al. (2012) con-
subunits, named CSN1–8 (Fig. 1A) (Bech-Otschir et al., 2005; firms a major interaction of CUL-1 with subunit CSN2 of the CSN
Cope and Deshaies, 2003; Deng et al., 2000; Harari-Steinberg and (Fig. 1B). The structural analysis of CSN–CRL complexes revealed a
Chamovitz, 2004; Kato and Yoneda-Kato, 2009; Schwechheimer, mutual interference of the two complexes. CSN-mediated dened-
2004; von Arnim, 2003; Wei and Deng, 2003; Wei et al., 2008; dylation causes a conformational change that deactivates the CRLs.
Wolf et al., 2003). The complex was originally discovered in Ara- On the other hand, CRL composition has impact on CSN-mediated
bidopsis thaliana as a regulator of constitutive photomorphogenesis deneddylation.
(COP) (Chamovitz et al., 1996; Wei et al., 1994). Subsequently it was
identified in human cells (Seeger et al., 1998) and studies using 3. Regulation of the CSN–CRL pathway
various eukaryotic organisms indicated that the CSN is responsi-
ble for regulating the UPS pathway (Glickman et al., 1998; Seeger The CSN–CRL pathway catalyze the specific ubiquitination of
et al., 1998; Wei and Deng, 1998). Deregulation of the CSN–CRL selected substrates which are subsequently degraded by the 26S
pathway can have dramatic effects on diverse cellular functions proteasome (Fig. 1A). Ubiquitination is stimulated by neddylation
including DNA repair (Groisman et al., 2003; Takedachi et al., 2010; and the removal of Nedd8 carried out by CSN5 through its met-
Wei et al., 2008), cell cycle control (Kato and Yoneda-Kato, 2009; alloenzyme (JAMM) domain (Cope et al., 2002) deactivates the
Liu et al., 2003), angiogenesis (Braumann et al., 2008) and tumor CRLs (Fig. 1B). In addition to this catalytic mechanism, CSN can
development as discussed in this review. also inhibit CRL function in a noncatalytic fashion (Emberley et al.,
2012; Enchev et al., 2012). The CSN-mediated inactivation of CRLs
2. Architecture of CSN–CRL supercomplexes prevents the autocatalytic breakdown and enables the reassem-
bly of the CRLs, maintaining their optimal activity (Schmidt et al.,
The CSN complex is composed of the six subunits with PCI 2009). The complex stability depends further on the availabil-
(Proteasome, COP9, and Initiation factor 3) domains (Fig. 1A, yel- ity of substrate (Bornstein et al., 2006; Chew and Hagen, 2007;
low subunits) (Aravind and Ponting, 1998; Glickman et al., 1998; Emberley et al., 2012). The presence of phosphorylated p27 e.g.
Hofmann and Bucher, 1998), and CSN5 and CSN6 with MPN (Mpr1p stimulates the activity of CRL1Skp2 and controls its own ubiquitin-
and Pad1p N-terminal) domains (Fig. 1A, blue subunits). The PCI ation and degradation (Bornstein et al., 2006). Also other CRLs were
and MPN domains are found almost exclusively in two other large found regulated by substrates (Chew and Hagen, 2007). CAND1 is
protein complexes, the 26S proteasome lid and the eukaryotic another important modulator of CRL activity maintenance in vivo. It
translation initiation factor eIF3, suggesting a common ancestor binds to the deneddylated form of cullins and competes with SRSs
in evolution (Pick et al., 2009). Initial structural studies of the (Zheng et al., 2002). Therefore, CAND1 allows SRS exchange by a
L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337 1329

Fig. 1. Model of the CSN–CRL pathway. (A) The COP9 signalosome (CSN) consists of eight subunits, six subunits possess PCI (Proteasome-COP9-Initiation factor-3) domains
(yellow) and two subunits MPN (Mpr1p and Pad1p N-terminal) domains (blue). The E3 cullin-RING ubiquitin ligase complex is composed of a scaffold cullin, RING box protein
(Rbx1), E2 ligase and a specific substrate recognition subunit (SRS). Neddylated CRL complexes ubiquitinate specific substrates (S) depending on their SRSs and promote
degradation by the ubiquitin proteasome system (UPS) via the 26S proteasome. (B) Deneddylation of the CRL by complex-bound CSN5 causes conformational changes and
shifts the CRL complex in an inactive state, which ensures complex stability and enables exchange of SRSs.

mechanism not yet fully understood (Emberley et al., 2012; (VHL) was reported proposing an interaction between these two
Goldenberg et al., 2004; Schmidt et al., 2009). SRSs (Roe et al., 2011). VHL, the SRS of the CRL2VHL complex, sta-
bilizes p27 and induces apoptosis in RCC cell lines and in vivo (Kim
4. Deregulation of the CSN–CRL pathway and its role in et al., 2004). Considering that Skp2 promotes the degradation of
tumorigenesis p27, Roe and coworkers revealed a VHL-mediated Skp2 polyubiqui-
tination and its degradation upon induction of DNA damage which
The fine-tuned regulation of the CSN–CRL pathway points to did not depend on the VHL E3 Ub ligase activity (Roe et al., 2011). In
an immense impact in case of disturbance. Thereby multiple com- bladder cancer, the Wnt pathway inhibitor WIF1 (Wnt inhibitory
ponents may be affected (Fig. 2A–D, Tables 1 and 2). Mutations factor 1) negatively regulates Skp2 by preventing the binding of
within substrate genes or additional post-transcriptional modifi- ␤-catenin to the Skp2 promotor, connecting the FBP to urothe-
cation prevent substrate recognition and promote accumulation lial tumorigenesis (Tang et al., 2009). Furthermore, deregulation of
of oncoproteins (Fig. 2A). Since SRSs play a fundamental role in Skp2 was found in prostate cancer (Lin et al., 2010; Yang et al., 2002)
proper functioning of CRLs, their loss or gain of function can lead (see Chapter 6.2). Another well-studied FBP, ␤-TrCP, behaves onco-
to cancerogenesis (Fig. 2C). Expression patterns of cullins, CSN sub- genic as well as tumor suppressive depending on cellular context.
units or CAND1 can be altered provoking oncogenesis. Moreover, ␤-TrCP targets cell cycle regulators leading to decreased genetic
tumor-viruses can abuse the cellular Ub machinery to manipulate instability (Guardavaccaro et al., 2003), although it was observed
the proteasome for their advantage (Fig. 2D). Examples mentioned upregulated in numerous tumors (Lau et al., 2012). Mutations and
below highlight the contribution of a deregulated CSN–CRL path- overexpression of ␤-TrCP were found in prostate cancer and pro-
way to tumorigenesis with focus on urological neoplasia. vided the first evidence for ␤-TrCP participation in tumorigenesis
(Gerstein et al., 2002). However, CRL1ˇ-TrCP ubiquitinates the two
4.1. Deregulation of substrate recognition subunits prominent oncoproteins ␤-catenin (Hart et al., 1999) and p53 ligase
Mdm2 (Inuzuka et al., 2010) impeding proliferation. Alterations in
The prominent FBP Skp2 was found overexpressed in almost the ␤-TrCP gene (BTRC) lead to accumulation of ␤-catenin and to
every tumor and correlates with poor prognosis fulfilling onco- development of colon cancer (Kim et al., 2007; Lau et al., 2012).
genic functions (Frescas and Pagano, 2008). Its expression is also Furthermore, a mutation was observed in the F-box motif of BTRC
increased in renal cell carcinoma (RCC), a tumor often accompanied induced tumorigenesis in murine kidneys through overexpression
with resistance to chemotherapy and kinase inhibitors like mam- and accumulation of ␤-catenin highlighting an anti-cancerogenic
malian target of rapamycin (mTOR) inhibitors (Langner et al., 2004; property of the FBP (Belaïdouni et al., 2005). Nevertheless, thera-
Liu et al., 2008). Interestingly, a correlation between mTOR inhibitor peutic targeting of ␤-TrCP might be intricate since its participation
sensitivity and Skp2 levels was detected suggesting an impact of in fundamental processes is tissue-dependent.
the FBP on therapy outcome (Totary-Jain et al., 2012) (see Chap- The FBP Fbw7 represents a classical tumor suppressor. CRL1Fbw7
ter 6.2). The downregulation of Skp2 by von Hippel–Lindau protein promotes degradation of numerous of tumor-driving proteins, and
1330 L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337

Fig. 2. The deregulated CSN–CRL pathway participates in tumorigenesis. Alterations of the CSN–CRL pathway caused by different modifications promote accumulation of
oncoproteins or increased degradation of tumor suppressors. (A) A substrate altered either through mutation or additional posttranslational modification avoids recognition
by the SRS of CRLs and therefore, accumulates in the cell. (B) Altered co-receptors impede the recognition of substrate. For example, an alteration in Cks1, the co-receptor
of Skp2 disturbs the recognition of p27 for ubiquitination (Frescas and Pagano, 2008). (C) Overexpressed SRS promotes enhanced tumor suppressor degradation whereas
downregulated or mutated SRS allows accumulation of oncoproteins. (D) Tumor-viruses are able to hijack the CRLs through SRS exchange with viral protein, interference
with the CRL core complex or deactivation by viral deneddylases.

mutation in Fbw7 substrate-binding regions or loss of heterozy- et al., 2006). Taken together, altered SRSs provide a platform for
gosity was found in 6% of all cancers, mainly lymphomas (Welcker drug interference. However, it still remains a challenge to iden-
and Clurman, 2008). The FBW7 gene is a direct target of p53, tify all substrates of SRSs in order to understand their complex
another well characterized tumor suppressor, indicating an anti- regulation function in a time and tissue-specific manner.
cancerogenic role (Mao et al., 2004). FBW7 gene mutations were
also identified in urological cancers (Liu et al., 2012). Consider- 4.2. Deregulation of cullins, CAND1 and CSN subunits
ing the Fbw7 substrates mTOR and Notch loss of function of this
FBP might play an important role in RCC development (Lau et al., Altered expression patterns of cullins, CSN subunits and CAND1
2012). Moreover, loss of Fbw7 promotes resistance to anti-tubulin were observed in various tumors. Berthold et al. reported a strong
chemotherapeutics like Taxol (Wertz et al., 2011), which may downregulation of CUL-3 in kidney cancer samples (Berthold et al.,
explain the leaking effect of these agents in RCC patients. 2008), whereas CUL-1 is upregulated in human lung tumors. Inter-
CRL1Fbw4 (containing an FBP related to Fbw7) impedes tumori- estingly, the neddylated form of CUL-1 is particularly expressed
genesis by ubiquitinating cyclin D1. Mutations that inhibit in high grade neuroendocrine lung tumors and associated with
dimerization of Fbw4 required for cyclin D1 degradation was found low levels of CAND1 (Salon et al., 2007). CUL-4A also plays a role
in esophageal carcinomas (Barbash et al., 2008). Keap1 (Kelch- in oncogenesis; the CUL-4A gene was found to be amplified or
like ECH-associated protein 1), another SRS protein, targets Nrf2 overexpressed in primary breast cancer and is associated with
(Nuclear factor (erythroid-derived 2)-like 2) to prevent its accu- aggressive growth and poor prognosis (Chen et al., 1998; Schindl
mulation and overexpression of Nrf2 downstream genes often et al., 2007). CUL-4A may contribute to tumorigenesis by promoting
observed in tumors. Its anti-oncogenic role is emphasized by 19% mTOR signaling, a key pathway in RCC, through degradation of the
of lung cancer patients harboring a somatic Keap1 mutation (Singh mTOR inhibitor REDD1 (Katiyar et al., 2009; Lee and Zhou, 2010).
L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337 1331

Table 1
Known deregulated components of the CSN–CRL pathway involved in tumorigenesis.

Target Group Deregulation Cancer Reference

Fbw4 SRS (FBP) Mutation prevents dimerization of Fbw4 and thus cyclin D1 Esophageal carcinomas Barbash et al. (2008)
degradation
Fbw7 SRS (FBP) Tumor suppressor, mutations in Fbw7 substrate-binding regions Lymphomas, multiple cancers Mao et al. (2004), Welcker and
Clurman (2008)
␤-TrCP SRS (FBP) Oncogenic/tumor suppressor, increases genetic instability, targets Multiple cancers Kim et al. (2007), Lau et al.
Mdm2, ␤-catenin (2012)
Skp2 SRS (FBP) Overexpression in multiple tumors, promotes degradation of p21, Multiple cancers Frescas and Pagano (2008)
p27
Keap1 SRS (BTB) Keap1 gene mutation drives Nrf2 accumulation, expression of Nrf2 Lung cancer Singh et al. (2006)
target genes
VHL SRS VHL gene negatively mutated or silenced, drives expression of Clear cell RCC Kim and Kaelin (2006)
HIF1␣ target genes
CUL-1 Cullins Expression levels increased, associated with poor prognosis, drives Melanoma, breast cancer Chen and Li (2010)
p27 degradation
CUL-3 Cullins Expression levels strongly decreased RCC samples Berthold et al. (2008)
CUL-4 Cullins CUL-4A gene amplified, overexpressed, protects mTOR through Breast cancer Chen et al. (1998), Katiyar et al.
degradation of REDD1 (2009)
CUL-5 Cullins Expression levels decreased Lung cancer Singhal et al. (2003)
CSN2 CSN CSN2 gene lost in several tumors, CSN2 over-expression drives Multiple cancers Leal et al. (2008)
chromosome instability
CSN3 CSN CSN3 gene amplified Osteosacroma Yan et al. (2007)
CSN5 CSN CSN5 overexpression in multiple tumors, CN5 gene amplified in Multiple cancers Patil et al. (2005), Shackleford
hepatocellular carcinoma and Claret (2010)
CSN6 CSN CSN6 overexpressed, stabilizes Mdm2 and promotes p53 Breast cancer, multiple cancer Lee et al. (2011b), Zhao et al.
degradation (2011)
CAND1 Expression levels decreased Prostate cancer Korzeniewski et al. (2012),
Murata et al. (2010)

Furthermore, CUL-5 is slightly upregulated in lung cancer shown the tumor suppressors p53 (Bech-Otschir et al., 2001) and p27
by expression profiling as it was also demonstrated for additional (Huang et al., 2006). In addition, upregulated CSN causes deregu-
cell cycle genes (Singhal et al., 2003). lation of the CSN–CRL pathway. Unfortunately, only little is known
CAND1 expression levels are decreased in prostate cancer about the function of the CSN in urological cancers so far. Since
(Korzeniewski et al., 2012; Murata et al., 2010). Since it is respon- CRL-bound FBPs are protected by the CSN-mediated deneddylation
sible for proper CRL function by promoting FBP exchange, CAND1 (Cope and Deshaies, 2006; Schmidt et al., 2009) it is likely that pri-
defects should also be considered important for prostate neoplasia mary modifications of the CSN cause alterations of FBPs resulting
development. in tumorigenesis.
The deregulation of pleiotropic functions of the CSN can also
participate in tumor development (Richardson and Zundel, 2005). 4.3. Impact of tumor-viruses on the CSN–CRL pathway
The CSN2 gene is lost in several types of cancer (Leal et al., 2008).
Amplification of the CSN3 gene in osteosarcomas indicates an onco- Tumor-viruses are able to hijack cellular Ub ligases (Fig. 2D,
genic role of the CSN subunit (Yan et al., 2007). CSN3 gene alteration Table 2) (Barry and Früh, 2006; Lee and Zhou, 2010). The HPV
is linked to the non-cancerous cerebric malignancy Smith–Magenis (human papilloma virus) E7 oncoprotein associates with the CUL-2
syndrome (Elsea et al., 1999). The CSN5 gene was found to be ampli- complex to CRL2E7 and redirects it to target the tumor suppressor
fied in hepatocellular carcinomas elucidating the high expression retinoblastoma protein contributing to the development of cervi-
levels in these tumors (Patil et al., 2005). CSN5 overexpression cal carcinoma (Huh et al., 2007). Moreover, the adenovirus-derived
correlated with tumor growth and negative clinical outcome was oncoprotein E1A inhibits CRL1Fbw7 by direct interaction with Rbx1
detected in numerous cancers (Shackleford and Claret, 2010). CSN6 and CUL-1 and promotes proliferation (Isobe et al., 2009). A nega-
gene amplification was observed in breast cancer followed by CSN6 tive regulation of CRL activity through a deneddylase encoded by
overexpression and Mdm2 stabilization (Zhao et al., 2011). How- Epstein–Barr virus, a tumor-virus, was found in lymphomas and
ever, it should be stressed out that most studies reporting elevated stomach cancer (Gastaldello et al., 2010). The same group revealed
CSN5 or CSN6 expression levels failed to give further informa- a further viral modulation of CRL activity by preventing the recruit-
tion on the amounts of other CSN subunit. Therefore it cannot be ment of CAND1, which can be rescued by CAND1 overexpression
excluded that the whole CSN complex rather than a single sub- (Gastaldello et al., 2012). In addition, the Kaposi’s sarcoma (KSH)-
unit is upregulated since convincing evidence for the independent virus protein latency-associated nuclear antigen (LANA) serves as
function of single CSN subunits is still lacking. A high cellular CSN a SRS in CRL5LANA and mediates the degradation of VHL and p53 to
level can induce cancer, because it accelerates the degradation of promote KSH development, one of the first recognized HIV-related

Table 2
Tumor-viruses attack the CSN–CRL pathway and promote tumorigenesis.

Tumor-virus protein Function Cancer Reference

Human papilloma virus E7 Associates with CRL2 core complex, drives retino-blastoma degradation Cervix carcinoma Huh et al. (2007), White et al.
(2012)
KSH-virus LANA Associates with CRL5 core complex, drives VHL, p53 degradation Kaposi’s sarcoma Cai et al. (2006)
Epstein–Barr-virus BPLF1 Viral deneddylase hydrolyses Nedd8 and stabilizes CRL substrates Lymphoma, stomach cancer Gastaldello et al. (2010)
Epstein–Barr-virus BZLF1 Associates with CRL2 or CRL5 core complex, promotes p53 degradation Lymphoma, stomach cancer Sato et al. (2009)
Hepatitis B virus Hbx Binds to Skp2 F-box region and protects c-Myc from degradation Hepatocellular carcinoma Kalra and Kumar (2006)
SV40 Large T antigen Interacts with Fbw7 and protects cyclin E from degradation Multiple tumors Welcker and Clurman (2005)
1332 L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337

Fig. 3. Regulation of the CSN–CRL pathway by published miRNAs. Different angles of the CSN–CRL pathway are affected by deregulated miRNAs. They either reduce (block
arrow) or indirectly induce the protein expression (arrow). The particular function of each miRNA is listed in Table 3.

illnesses (Cai et al., 2006). These examples illustrate that tumor- miR-146a in gastric cancer (Crone et al., 2012). Like CSN subunits,
viruses target the CSN–CRL pathway to use it for their own purpose, also E2 ligase Cdc34 mRNA is regulated by the let-7 family. Low
providing another example for the significance of the pathway in Cdc34 levels due to let-7b overexpression resulted in decreased
cancer development. CRL1 activity, stabilizing Wee1 and G2 /M accumulation (Legesse-
Miller et al., 2009). Furthermore, CAND1 was spotted as a target of
5. miRNA-mediated regulation of the CSN–CRL pathway androgen-responsive miR-148a in prostate cancer (Murata et al.,
2010).
5.1. Impact of the miRNome on the CSN–CRL complex Besides CSN and CAND1 also cullins and SRSs are negatively reg-
ulated by miRNAs. The hypoxia-sensitive miR-424 decreases CUL-2
The enormous influx of novel miRNA studies in public databases levels and promotes angiogenesis, a phenotype that often occurs in
point out the significance of miRNAs in tumor development, in RCC (Ghosh et al., 2010). Targeting of CUL-5 by miR-19a/19b was
monitoring tumor progression and as possible therapeutic tar- reported in cervical carcinoma cells (Xu et al., 2012). The miR-19a
gets. Deregulation within the miRNA collectivity (miRNome) has and miR-19b belong to the miR-17-92 cluster and were demon-
a major impact on the equilibrium of the proteome in cells on a strated to be strongly oncogenic by targeting E2Fs and by promoting
post-transcriptional level often leading to pathological conditions. overexpression of the MYC gene in many tumors. Paradoxically,
On the other hand, post-translational modification e.g. ubiquitin- besides the oncogenic characteristics, this cluster also shows tumor
ation can lead to fine-tuned protein degradation and physiological suppressor features which need to be further inspected (Zhang
maintenance. Both fundamental processes play an important role et al., 2009). The strongest targeted SRS remained the tumor sup-
in tumorigenesis. But how does the miRNome interact with the pressor protein Fbw7. Its mRNA is negatively regulated by miR-25,
protein degradation machinery? Here we examine whether dereg- -27a and -223 in different contexts (Lerner et al., 2011). Increased
ulated miRNAs could also affect the UPS with focus on the CSN–CRL genome instability triggered by miR-223 targeting Fbw7 provided
pathway. Fig. 3 displays an overview on known miRNAs affecting the first evidence that CRL1s are affected by the miRNome (Xu et al.,
the ubiquitination mechanism. Furthermore, the function of these 2010). In addition, FBP ␤-TrCP is targeted by miRNAs. The miR-183
particular miRNAs and their deregulation in cancer is provided in interacts with ␤-TrCP mRNA, a mechanism that can be prevented
Table 3. by binding of specific proteins (Elcheva et al., 2009). The inves-
The regulation of CSN subunit expression by the let-7 miRNA tigation of the correlation of miRNAs and cisplatin resistance in
family was first discovered in the context of CSN assembly in 2011 ovarian cancers revealed a direct regulation of Keap1 by miR-141
(Leppert et al., 2011). In addition, the subunit CSN8 is targeted by (van Jaarsveld et al., 2012). The miR-141 also plays a critical role
L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337 1333

Table 3
Regulation of the CSN–CRL pathway by miRNAs.

Target miRNA Function Tissue References

CSN miR-Let-7 Targets CSN subunit mRNAs, inhibits assembly Cervical cancer Leppert et al. (2011)
CSN8 miR-146a Targets CSN8, inhibits NF␬B activation Gastric cancer (mouse model) Crone et al. (2012)
CAND1 miR-148a Negative regulation of CAND1 expression Prostate cancer Murata et al. (2010)
Cdc34 miR-Let-7 Targets Cdc34 expression, promotes cell cycle arrest Primary human fibroblasts Legesse-Miller et al. (2009)
CUL-2 miR-424 Targets CUL-2, promotes angiogenesis Cervical cancer Ghosh et al. (2010)
CUL-5 miR-19a, 19b Negative regulation of CUL-5 expression Cervical cancer Xu et al. (2012)
Fbw7 miR-223 Targets Fbxw7, promotes genomic instability Embryonic fibroblasts Li et al. (2012), Xu et al. (2010)
miR-27a Targets Fbxw7, promotes cell cycle progression Various cancer cell lines Lerner et al. (2011)
miR-25 Targets Fbxw7 Embryonic fibroblasts Lu et al. (2012)
␤-TrCP miR-183 Negative regulation of ␤-TrCP expression Colorectal cancer cell lines Elcheva et al. (2009)
VHL miR-92a Negative regulation of VHL expression RCC samples Valera et al. (2011)
Keap1 miR-141 Targets Keap1, induces cisplatin resistance Ovarian cancer van Jaarsveld et al. (2012)
p27 miR-221 miR-222 Negative regulation of p27 expression Cardiomyocytes Wang et al. (2012b)
miR-181 Negative regulation of p27 expression Hepatic stellate cells Wang et al. (2012a)
p53 miR-214 Indirect upregulation of p53 Myeloma cell lines Misiewicz-Krzeminska et al. (2012)
c-Myc miR-34a Targets c-Myc, inhibits invasion Prostate cancer Yamamura et al. (2012)
FOXO1 miR-27a, miR-96,-182 Negative regulation of FOXO1 expression Breast cancer Guttilla and White (2009)
c-Jun miR-125b Negative regulation of c-Jun expression Melanoma cell lines, samples Kappelmann et al. (2012)
miR-155 Negative regulation of c-Jun expression Human dermal fibroblasts Song et al. (2012)
Cyclin D1 miR-520b Negative regulation of cyclin D1 and MEKK2 expression Hepatoma cell lines Zhang et al. (2012)

in urological cancers. The VHL protein is under the control of miR- anti-neddylation agent MLN4924 strikes by inhibiting complete
92a leading to stabilization of pro-angiogenic factors (Valera et al., CRL activity (Soucy et al., 2010) and is only useful when an over-
2011). It should be mentioned that substrates of CRLs are regulated expressed cullin or FBP is the main key player in a specific cancer
by miRNAs as well (Table 3). type. Nevertheless, radio sensitization of cancer cells was found
This data gives an insight in the regulation of the CSN–CRL path- after inhibition of CRLs, which could provide an advantage for
way by the miRNome. Different angles of the Ub machinery are tumor patients (Wei et al., 2012). In order to reduce adverse reac-
affected by deregulated miRNAs in various tumors (Fig. 3, Table 3). tion, more specific drugs are needed which interfere with substrate
Interestingly, most of these miRNAs are also altered in urological recognition or required kinases like the cyclin-dependent kinase
cancers providing a new regulation cross-talk and possible com- regulatory subunit (Cks1) for Skp2-mediated p27 recognition. The
mon targets for these tumors. small molecule enhancer of rapamycin (SMER3) is able to inhibit
the binding of FBP Met30 to the CRL1 core complex (Aghajan et al.,
5.2. Evaluated miRNAs altered in urological cancers and their 2010) and is an example for such a unique interference with the
possible impact on components of the CSN–CRL pathway CSN–CRL pathway.

Insufficient data were published so far concerning urological 6.2. The CSN–CRL pathway component Skp2 might be a specific
cancer-associated miRNAs and the impact of the CSN–CRL complex. therapeutic target in urological cancers
To identify putative miRNA binding sites in the mRNAs of members
of the CSN–CRL complex, an in silico target prediction using Tar- Urological cancers are accompanied by increasing incidence and
getScan 6.2 algorithm was performed, a stringent tool to identify poor prognosis longing for a better understanding of molecular
miRNA binding sites (http://www.targetscan.org/). Table 4 summa- mechanisms and new therapeutic targets. In RCC, the therapeutic
rizes predicted targeting of components of the CSN–CRL pathway opportunities are limited since RCC patients are mostly resistant to
by selected miRNAs found to be deregulated in urological neoplasia chemo- and radiation therapy (Corn, 2007) and they only partly
(Schaefer et al., 2010; Wotschofsky et al., 2012). The CSN, CAND1, respond to currently available agents. The proteasome inhibitor
cullins and FBPs were spotted as predicted targets of these partic- bortezomib was found in a phase II clinical trial with only par-
ular deregulated miRNAs. Especially CSN2, CSN7b, CUL-3, CUL-4, tial response in 10% of RCC patients (Kondagunta et al., 2004).
␤-TrCP and FBPs containing a WD40 domain (FBXWs) provide puta- The toxicity caused by the agent recommend inhibition of E3
tive miRNA binding sites, which need to be verified in experimental Ub ligases as a more specific treatment (Corn, 2007). Totary-Jain
context. Based on this prediction, a urological cancer-promoting et al. revealed an interesting interplay between the mTOR inhibitor
miRNome seems to play a role in the regulation of the CSN–CRL rapamycin and Skp2. Rapamycin down-regulates Skp2 by impair-
pathway (Table 4) and should be considered in future investigations ing its phosphorylation through AKT (Protein kinase B) and RNA
of tumor therapeutics. interference-mediated silencing of Skp2 increased sensitivity of
tumor cells to rapamycin in vitro and in vivo (Totary-Jain et al.,
6. Is the CSN–CRL pathway a suitable target for cancer 2012). Therefore, the Skp2 expression level can serve as a pre-
therapy? dictor for mTOR inhibitor-sensitivity. For RCC tumors resistant to
mTOR inhibitors, additional targeting of Skp2 might be a ther-
6.1. The deregulated CSN–CRL pathway as a target in cancer apeutic option. In recent years, Skp2 emerged as an attractive
therapy target in prostate cancer therapy. Evidence of Skp2 overexpress-
ion in neoplastic prostate tissue accumulates (Lin et al., 2010;
The deregulated CSN–CRL pathway is an attractive target for Shim et al., 2003; Yang et al., 2002; Zheng et al., 2004), correlating
cancer therapy. This approach will improve and complement ther- with tumor grade and poor prognosis like in most types of cancer
apies already developed in the past decade. Proteasome inhibitors (Frescas and Pagano, 2008). Using high-throughput screening, the
effectively downregulate protein degradation but cause a vari- small molecule inhibitor CpdA (Compound A) was identified which
ety of unwanted side effects and are only approved for specific blocks the recruitment of Skp2 to the CRL core complex (Chen et al.,
types of myeloma and lymphoma (D’Arcy and Linder, 2012). The 2008). CpdA was able to overcome the resistance to chemotherapy
1334 L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337

Table 4
Predicted targets of deregulated miRNAs in prostate and renal cell cancer (Schaefer et al., 2010; Wotschofsky et al., 2012) with focus on the CSN–CRL pathway.

Target Gene Protein Kidney cancer Prostate cancer

CSN COPS2 CSN2 miR-16,-31,-32,-92a,-92b, miR-107,-181b


COPS7A CSN7a miR-16
COPS7B CSN7b miR-10 miR-16,-34a, -34a,c,-125, miR-486-5p
COPS8 CSN8 miR-200c
CAND1 CAND1 CAND1 miR-148
Cullins CUL2 CUL-2 miR-16,-574-3p
CUL3 CUL-3 miR-130a miR-20a,-23b,-32,-34b, miR-92a,-92b,-101,-106a,
miR-106b,-141,-181b,-218, miR-373, -485-3p,-636
CUL4A CUL-4a miR-514 miR-96,-107,-181b
CUL4B CUL-4b miR-101,-141,-194
CUL5 CUL-5 miR-19a,-19b miR-31,-96,-101,-145,-182, miR-181b
F-box proteins BTRC ␤-TrCP miR-10b, -195, miR-224, -514 miR-16,-34b,135b,-107
SKP1 Skp1 miR-221,-145
FBXWs FBXW2 Fbw2 miR-29a,-141,-146a, -194, miR-197,-331-3p
FBXW4 Fbw4 miR-125b,-149
FBXW5 Fbw5 miR-296
FBXW7 Fbw7 miR-223 miR-1,-16,-29a,-32,-92a, miR-92b,-101,-107,-194, miR-197
FBW9 Fbw9 miR-29b miR-29a
FBXW11 Fbw11 miR-20a,-101, miR-200c miR-20a,-23b,-96,-101, miR-181b,-182

and proteasome inhibitors in multiple myeloma, highlighting the known about the regulation of the CSN–CRL pathway by miRNAs
potential role of targeting Skp2 in the battle against tumor progres- and by signaling pathways, and research in this field might open a
sion (Chen et al., 2008). Moreover, natural occurring compounds new area of pharmacological intervention.
like curcumin, vitamin D and others promoted the downregulation Which therapeutics is desirable and applicable in cancer
of Skp2 inducing p27 stabilization (Huang et al., 2011; Yang and therapy?
Burnstein, 2003). The novel energy restriction-mimic agent CG- In the past decade progress has been made in the develop-
12 also downregulates Skp2 and induces ␤-TrCP upregulation by ment of drugs interfering with the UPS. Proteasome inhibitors
preventing the Skp2-␤-TrCP-Sp1-feedback loop, a novel cross-talk prevent degradation of essential tumor suppressors. However,
found between these two important FBPs (Wei et al., 2012). Never- overall blocking of the proteasome leads to severe side effects,
theless, the development of Skp2 therapeutics is still accompanied because of the pleiotropic role of the enzyme complex. Similar
by obstructions due to a lack of appropriate delivery systems. The outcome can be expected with the use of neddylation as well as
Skp2 substrate recognition of both tumor suppressors and onco- CSN-mediated deneddylation inhibitors, since the function of all
proteins might also complicate their usage. However, Skp2 forms a CRLs will be impeded. To reduce adverse reactions more specific
structurally active site in the presence of co-receptor kinase Cks1 drugs are needed which interfere with particular SRSs or prevent
(Fig. 2B) resulting in specific ubiquitination of tumor suppressor tumor suppressor recognition and elimination. RNAi-based agents
substrates like p27 (Huang et al., 2012). Drugs interfering with the are imaginable to stop the biosynthesis of a particular cullin or
Skp2-Cks1 binding site or Skp2-p27 interface might be attractive SRS. Moreover, engineering specific SRS that target oncoproteins
therapeutic agents which exclusively prevent nuclear p27 degra- for degradation are up for debate. The potential involvement of
dation associated with tumor development (Cardozo and Pagano, deregulated miRNAs should also be considered. The determination
2007). of miRNA levels emerged as powerful diagnostic and prognostic
tool in urological cancers. miRNAs are stable, and non-invasive
7. Future prospects detection is possible. Here, we provide evidence that an altered
miRNome influences the CSN–CRL pathway and, therefore, could
The CSN–CRL pathway is dependent on the elaborate interplay be an attractive target for further examinations.
between enzymatic protein complexes. Whereas the CSN influ- What is the role of the CSN–CRL pathway in urological
ences CRL activity mostly by deneddylation, CRL components and tumorigenesis?
substrates modulate CSN-mediated deneddylation. The pathway New applicable targets are needed for the treatment of uro-
confers substrate specificity to the UPS and is virtually responsible logical cancer. Indeed, altered expression of important FBPs in
for the degradation of a specific protein, at a certain time in a par- urological cancers influences the presence of key tumor pro-
ticular compartment. Because oncoproteins and tumor suppressors moting proteins in urological tissues as reported in this review.
are among the substrates of the CSN–CRL complex, deregulation The exact mechanism of how deregulated SRSs are integrated
of its parts impairs intracellular protein dynamics with the preva- in urological tumorigenesis is currently unknown. Unfortunately,
lent consequence of tumor development as outlined in this review. clinical data of CSN subunits in urological cancer samples are still
Because modifications of the CSN–CRL pathway are increasingly lacking. Therefore, for the further understanding and therapy of
found in cancers, the pathway becomes an interesting field for urological tumors, the CSN–CRL pathway would be an exciting
pharmacological intervention in cancer. subject.
What are relevant open questions in our understanding of How can gained knowledge be transferred to cancer therapy
molecular mechanisms of the CSN–CRL pathway? development?
Future investigations should focus on concrete mechanisms and Pre-clinical results reviewed here clearly recommend the
molecular structures of the pathway. At the moment the struc- CSN–CRL pathway as a major target for future tumor therapy.
tural model of the CSN–CRL complex is a prediction and has to be Compounds with clear anti-tumor evidence in cell culture and in
verified by cryo-electron microscopy and crystallography. A com- animals should be advanced in clinical trials. In the near future,
prehensive characterization of SRSs and their substrates and their well designed clinical studies should to be carried out to assess the
deregulation in cancer is a prerequisite for the development of clinical properties and application of possible new drugs that target
specific compounds targeting SRS/substrate interactions. Little is the CSN–CRL pathway.
L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337 1335

Acknowledgments Elcheva I, Goswami S, Noubissi FK, Spiegelman VS. CRD-BP protects the coding
region of betaTrCP1 mRNA from miR-183-mediated degradation. Molecular Cell
2009;35:240–6.
This work was funded by the Foundation of Urological Research, Elsea SH, Mykytyn K, Ferrell K, Coulter KL, Das P, Dubiel W, et al. Hemizygosity for
Berlin and the Charité-Universitätsmedizin, Berlin. Due to space the COP9 signalosome subunit gene, SGN3, in the Smith-Magenis syndrome.
limitations we could not consider each article of the field in this American Journal of Medical Genetics 1999;87:342–8.
Emberley ED, Mosadeghi R, Deshaies RJ. Deconjugation of Nedd8 from Cul1 is
review. We apologize for that incompleteness. directly regulated by Skp1-F-box and substrate, and the COP9 signalosome
inhibits deneddylated SCF by a noncatalytic mechanism. Journal of Biological
Chemistry 2012;287:29679–89.
References Enchev RI, Schreiber A, Beuron F, Morris EP. Structural insights into the COP9 sig-
nalosome and its common architecture with the 26S proteasome lid and eIF3.
Aghajan M, Jonai N, Flick K, Fu F, Luo M, Cai X, et al. Chemical genetics screen Structure 2010;18:518–27.
for enhancers of rapamycin identifies a specific inhibitor of an SCF family E3 Enchev RI, Scott DC, da Fonseca PC, Schreiber A, Monda JK, Schulman BA, et al.
ubiquitin ligase. Nat Biotechnol 2010;28:738–42. Structural basis for a reciprocal regulation between SCF and CSN. Cell Reports
Aravind L, Ponting CP. Homologues of 26S proteasome subunits are regulators of 2012;2:616–27.
transcription and translation. Protein Science 1998;7:1250–4. Frescas D, Pagano M. Deregulated proteolysis by the F-box proteins SKP2 and beta-
Barbash O, Zamfirova P, Lin DI, Chen X, Yang K, Nakagawa H, et al. Mutations in TrCP: tipping the scales of cancer. Nature Reviews Cancer 2008;8:438–49.
Fbx4 inhibit dimerization of the SCF(Fbx4) ligase and contribute to cyclin D1 Gastaldello S, Callegari S, Coppotelli G, Hildebrand S, Song M, Masucci MG. Herpes
overexpression in human cancer. Cancer Cell 2008;14:68–78. virus deneddylases interrupt the cullin-RING ligase neddylation cycle by inhib-
Barry M, Früh K. Viral modulators of cullin RING ubiquitin ligases: culling the host iting the binding of CAND1. Journal of Molecular Cell Biology 2012;4:242–51.
defense. In: Sci STKE. 2006; 2006. p. pe21. Gastaldello S, Hildebrand S, Faridani O, Callegari S, Palmkvist M, Di Guglielmo C,
Bech-Otschir D, Kapelari B, Dubiel W. The COP9 signalosome: its possible role in the et al. A deneddylase encoded by Epstein-Barr virus promotes viral DNA repli-
ubiquitin system. In: Mayer JR, Ciechanover A, Rechsteiner M, editors. Ubiquitin cation by regulating the activity of cullin-RING ligases. Nature Cell Biology
and the chemistry of life. Weinheim: WILEY-VCH Verlag GmbH & KGaA; 2005. 2010;12:351–61.
p. 348–69. Gerstein AV, Almeida TA, Zhao G, Chess E, Shih IM, Buhler K, et al. APC/CTNNB1 (beta-
Bech-Otschir D, Kraft R, Huang X, Henklein P, Kapelari B, Pollmann C, et al. COP9 catenin) pathway alterations in human prostate cancers. Genes Chromosomes
signalosome-specific phosphorylation targets p53 to degradation by the ubi- Cancer 2002;34:9–16.
quitin system. EMBO Journal 2001;20:1630–9. Ghosh G, Subramanian IV, Adhikari N, Zhang X, Joshi HP, Basi D, et al. Hypoxia-
Belaïdouni N, Peuchmaur M, Perret C, Florentin A, Benarous R, Besnard-Guérin C. induced microRNA-424 expression in human endothelial cells regulates
Overexpression of human beta TrCP1 deleted of its F box induces tumorigenesis HIF-␣ isoforms and promotes angiogenesis. Journal of Clinical Investigation
in transgenic mice. Oncogene 2005;24:2271–6. 2010;120:4141–54.
Berthold J, Schenková K, Ramos S, Miura Y, Furukawa M, Aspenström P, et al. Char- Glickman MH, Rubin DM, Coux O, Wefes I, Pfeifer G, Cjeka Z, et al. A subcomplex of the
acterization of RhoBTB-dependent Cul3 ubiquitin ligase complexes—evidence proteasome regulatory particle required for ubiquitin-conjugate degradation
for an autoregulatory mechanism. Experimental Cell Research 2008;314: and related to the COP9-signalosome and eIF3. Cell 1998;94:615–23.
3453–65. Goldenberg SJ, Cascio TC, Shumway SD, Garbutt KC, Liu J, Xiong Y, et al. Structure of
Bondar T, Kalinina A, Khair L, Kopanja D, Nag A, Bagchi S, et al. Cul4A and DDB1 the Cand1-Cul1-Roc1 complex reveals regulatory mechanisms for the assembly
associate with Skp2 to target p27Kip1 for proteolysis involving the COP9 sig- of the multisubunit cullin-dependent ubiquitin ligases. Cell 2004;119:517–28.
nalosome. Molecular and Cellular Biology 2006;26:2531–9. Groisman R, Polanowska J, Kuraoka I, Sawada J, Saijo M, Drapkin R, et al. The ubiquitin
Bornstein G, Ganoth D, Hershko A. Regulation of neddylation and deneddyla- ligase activity in the DDB2 and CSA complexes is differentially regulated by the
tion of cullin1 in SCFSkp2 ubiquitin ligase by F-box protein and substrate. COP9 signalosome in response to DNA damage. Cell 2003;113:357–67.
Proceedings of the National Academy of Sciences of the United States of America Guardavaccaro D, Kudo Y, Boulaire J, Barchi M, Busino L, Donzelli M, et al. Con-
2006;103:11515–20. trol of meiotic and mitotic progression by the F box protein beta-Trcp1 in vivo.
Braumann C, Tangermann J, Jacobi CA, Müller JM, Dubiel W. Novel anti-angiogenic Developmental Cell 2003;4:799–812.
compounds for application in tumor therapy—COP9 signalosome-associated Guttilla IK, White BA. Coordinate regulation of FOXO1 by miR-27a, miR-96, and miR-
kinases as possible targets. Mini-Reviews in Medicinal Chemistry 2008;8: 182 in breast cancer cells. Journal of Biological Chemistry 2009;284:23204–16.
421–8. Harari-Steinberg O, Chamovitz DA. The COP9 signalosome: mediating between
Cai QL, Knight JS, Verma SC, Zald P, Robertson ES. EC5S ubiquitin complex is recruited kinase signaling and protein degradation. Current Protein & Peptide Science
by KSHV latent antigen LANA for degradation of the VHL and p53 tumor sup- 2004;5:185–9.
pressors. PLOS Pathogens 2006;2:e116. Hart M, Concordet JP, Lassot I, Albert I, del los Santos R, Durand H, et al. The F-box
Cardozo T, Pagano M. Wrenches in the works: drug discovery targeting the SCF protein beta-TrCP associates with phosphorylated beta-catenin and regulates
ubiquitin ligase and APC/C complexes. BMC Biochemistry 2007;8(Suppl. 1):S9. its activity in the cell. Current Biology 1999;9:207–10.
Chamovitz DA, Wei N, Osterlund MT, von Arnim AG, Staub JM, Matsui M, et al. The Hershko A, Ciechanover A. The ubiquitin system. Annual Review of Biochemistry
COP9 complex, a novel multisubunit nuclear regulator involved in light control 1998;67:425–79.
of a plant developmental switch. Cell 1996;86:115–21. Hofmann K, Bucher P. The PCI domain: a common theme in three multiprotein
Chen G, Li G. Increased Cul1 expression promotes melanoma cell prolifera- complexes. Trends in Biochemical Sciences 1998;23:204–5.
tion through regulating p27 expression. International Journal of Oncology Huang HC, Lin CL, Lin JK. 1,2,3,4,6-Penta-O-galloyl-␤-D-glucose, quercetin, curcumin
2010;37:1339–44. and lycopene induce cell-cycle arrest in MDA-MB-231 and BT474 cells through
Chen LC, Manjeshwar S, Lu Y, Moore D, Ljung BM, Kuo WL, et al. The human homo- downregulation of Skp2 protein. Journal of Agricultural and Food Chemistry
logue for the Caenorhabditis elegans cul-4 gene is amplified and overexpressed 2011;59:6765–75.
in primary breast cancers. Cancer Research 1998;58:3677–83. Huang KT, Pavlides SC, Lecanda J, Blank SV, Mittal KR, Gold LI. Estrogen and proges-
Chen Q, Xie W, Kuhn DJ, Voorhees PM, Lopez-Girona A, Mendy D, et al. Targeting terone regulate p27kip1 levels via the ubiquitin-proteasome system: pathogenic
the p27 E3 ligase SCF(Skp2) results in p27- and Skp2-mediated cell-cycle arrest and therapeutic implications for endometrial cancer. PLoS ONE 2012;7:e46072.
and activation of autophagy. Blood 2008;111:4690–9. Huang X, Langelotz C, Hetfeld-Pechoc BK, Schwenk W, Dubiel W. The COP9 sig-
Chew EH, Hagen T. Substrate-mediated regulation of cullin neddylation. Journal of nalosome mediates beta-catenin degradation by deneddylation and blocks
Biological Chemistry 2007;282:17032–40. adenomatous polyposis coli destruction via USP15. Journal of Molecular Biology
Cope GA, Deshaies RJ. COP9 signalosome: a multifunctional regulator of SCF and 2009;391:691–702.
other cullin-based ubiquitin ligases. Cell 2003;114:663–71. Huang X, Wagner E, Dumdey R, Peth A, Berse M, Dubiel W. Phosphorylation by
Cope GA, Deshaies RJ. Targeted silencing of Jab1/Csn5 in human cells downregu- COP9 signalosome-associated CK2 promotes degradation of p27 during the G1
lates SCF activity through reduction of F-box protein levels. BMC Biochemistry cell cycle phase. Israel Journal of Chemistry 2006;46:231–8.
2006;7:1. Huh K, Zhou X, Hayakawa H, Cho JY, Libermann TA, Jin J, et al. Human papillo-
Cope GA, Suh GS, Aravind L, Schwarz SE, Zipursky SL, Koonin EV, et al. Role of mavirus type 16 E7 oncoprotein associates with the cullin 2 ubiquitin ligase
predicted metalloprotease motif of Jab1/Csn5 in cleavage of Nedd8 from Cul1. complex, which contributes to degradation of the retinoblastoma tumor sup-
Science 2002;298:608–11. pressor. Journal of Virology 2007;81:9737–47.
Corn PG. Role of the ubiquitin proteasome system in renal cell carcinoma. BMC Inuzuka H, Tseng A, Gao D, Zhai B, Zhang Q, Shaik S, et al. Phosphorylation by casein
Biochemistry 2007;8(Suppl. 1):S4. kinase I promotes the turnover of the Mdm2 oncoprotein via the SCF(beta-TRCP)
Crone SG, Jacobsen A, Federspiel B, Bardram L, Krogh A, Lund AH, et al. microRNA- ubiquitin ligase. Cancer Cell 2010;18:147–59.
146a inhibits G protein-coupled receptor-mediated activation of NF-kappaB by Isobe T, Hattori T, Kitagawa K, Uchida C, Kotake Y, Kosugi I, et al. Adenovi-
targeting CARD10 and COPS8 in gastric cancer. Molecular Cancer 2012;11:71. rus E1A inhibits SCF(Fbw7) ubiquitin ligase. Journal of Biological Chemistry
D’Arcy P, Linder S. Proteasome deubiquitinases as novel targets for cancer therapy. 2009;284:27766–79.
International Journal of Biochemistry & Cell Biology 2012;44:1729–38. Kalra N, Kumar V. The X protein of hepatitis B virus binds to the F box protein Skp2
Deng XW, Dubiel W, Wei N, Hofmann K, Mundt K, Colicelli J, et al. Unified nomen- and inhibits the ubiquitination and proteasomal degradation of c-Myc. FEBS
clature for the COP9 signalosome and its subunits: an essential regulator of Letters 2006;580:431–6.
development. Trends in Genetics 2000;16:202–3. Kapelari B, Bech-Otschir D, Hegerl R, Schade R, Dumdey R, Dubiel W. Electron
Deshaies RJ, Joazeiro CA. RING domain E3 ubiquitin ligases. Annual Review of Bio- microscopy and subunit-subunit interaction studies reveal a first architecture
chemistry 2009;78:399–434. of COP9 signalosome. Journal of Molecular Biology 2000;300:1169–78.
1336 L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337

Kappelmann M, Kuphal S, Meister G, Vardimon L, Bosserhoff AK. MicroRNA miR- Peng Z, Shen Y, Feng S, Wang X, Chitteti B, Vierstra R, et al. Evidence for a physical
125b controls melanoma progression by direct regulation of c-Jun protein association of the COP9 signalosome, the proteasome, and specific SCF E3 ligases
expression. Oncogene 2012., http://dx.doi.org/10.1038/onc.2012.307. in vivo. Current Biology 2003;13:R504–5.
Katiyar S, Liu E, Knutzen CA, Lang ES, Lombardo CR, Sankar S, et al. REDD1, an Petroski MD, Deshaies RJ. Function and regulation of cullin-RING ubiquitin ligases.
inhibitor of mTOR signalling, is regulated by the CUL4A-DDB1 ubiquitin ligase. Nature Reviews Molecular Cell Biology 2005;6:9–20.
EMBO Reports 2009;10:866–72. Pick E, Hofmann K, Glickman MH. PCI complexes: Beyond the proteasome, CSN, and
Kato JY, Yoneda-Kato N. Mammalian COP9 signalosome. Genes Cells eIF3 Troika. Molecular Cell 2009;35:260–4.
2009;14:1209–25. Richardson KS, Zundel W. The emerging role of the COP9 signalosome in cancer.
Kim CJ, Song JH, Cho YG, Kim YS, Kim SY, Nam SW, et al. Somatic mutations of the Molecular Cancer Research 2005;3:645–53.
beta-TrCP gene in gastric cancer. APMIS 2007;115:127–33. Roe JS, Kim HR, Hwang IY, Cho EJ, Youn HD. von Hippel-Lindau protein promotes
Kim M, Yan Y, Lee K, Sgagias M, Cowan KH. Ectopic expression of von Hippel- Skp2 destabilization on DNA damage. Oncogene 2011;30:3127–38.
Lindau tumor suppressor induces apoptosis in 786-O renal cell carcinoma cells Saha A, Deshaies RJ. Multimodal activation of the ubiquitin ligase SCF by Nedd8
and regresses tumor growth of 786-O cells in nude mouse. Biochemical and conjugation. Molecular Cell 2008;32:21–31.
Biophysical Research Communications 2004;320:945–50. Salon C, Brambilla E, Brambilla C, Lantuejoul S, Gazzeri S, Eymin B. Altered pattern of
Kim WY, Kaelin WG. Molecular pathways in renal cell carcinoma—rationale for Cul-1 protein expression and neddylation in human lung tumours: relationships
targeted treatment. Seminars in Oncology 2006;33:588–95. with CAND1 and cyclin E protein levels. Journal of Pathology 2007;213:303–10.
Kondagunta G, Drucker B, Schwartz L, Bacik J, Marion S, Russo P, et al. Phase II trial of Sato Y, Kamura T, Shirata N, Murata T, Kudoh A, Iwahori S, et al. Degradation of
bortezomib for patients with advanced renal cell carcinoma. Journal of Clinical phosphorylated p53 by viral protein-ECS E3 ligase complex. PLOS Pathogens
Oncology 2004;22:3720–5. 2009;5:e1000530.
Korzeniewski N, Hohenfellner M, Duensing S. CAND1 promotes PLK4-mediated cen- Schaefer A, Stephan C, Busch J, Yousef GM, Jung K. Diagnostic, prognostic and ther-
triole overduplication and is frequently disrupted in prostate cancer. Neoplasia apeutic implications of microRNAs in urologic tumors. Nature Reviews Urology
2012;14:799–806. 2010;7:286–97.
Langner C, von Wasielewski R, Ratschek M, Rehak P, Zigeuner R. Biological signifi- Schindl M, Gnant M, Schoppmann SF, Horvat R, Birner P. Overexpression of the
cance of p27 and Skp2 expression in renal cell carcinoma. A systematic analysis human homologue for Caenorhabditis elegans cul-4 gene is associated with
of primary and metastatic tumour tissues using a tissue microarray technique. poor outcome in node-negative breast cancer. Anticancer Research 2007;27:
Virchows Archiv 2004;445:631–6. 949–52.
Lau AW, Fukushima H, Wei W. The Fbw7 and betaTRCP E3 ubiquitin ligases and their Schmidt MW, McQuary PR, Wee S, Hofmann K, Wolf DA. F-box-directed CRL
roles in tumorigenesis. Frontiers in Bioscience 2012;17:2197–212. complex assembly and regulation by the CSN and CAND1. Molecular Cell
Leal JF, Fominaya J, Cascón A, Guijarro MV, Blanco-Aparicio C, Lleonart M, et al. Cel- 2009;35:586–97.
lular senescence bypass screen identifies new putative tumor suppressor genes. Schwechheimer C. The COP9 signalosome (CSN): an evolutionary conserved pro-
Oncogene 2008;27:1961–70. teolysis regulator in eukaryotic development. Biochimica et Biophysica Acta
Lee J, Zhou P. Cullins and cancer. Genes Cancer 2010;1:690–9. 2004;1695:45–54.
Lee JE, Sweredoski MJ, Graham RL, Kolawa NJ, Smith GT, Hess S, et al. The steady-state Schwechheimer C, Serino G, Callis J, Crosby WL, Lyapina S, Deshaies RJ, et al. Interac-
repertoire of human SCF ubiquitin ligase complexes does not require ongoing tions of the COP9 signalosome with the E3 ubiquitin ligase SCFTIRI in mediating
Nedd8 conjugation. Molecular & Cellular Proteomics 2011a;10:M110:006460. auxin response. Science 2001;292:1379–82.
Lee MH, Zhao R, Phan L, Yeung SC. Roles of COP9 signalosome in cancer. Cell Cycle Seeger M, Kraft R, Ferrell K, Bech-Otschir D, Dumdey R, Schade R, et al. A novel
2011b;10:3057–66. protein complex involved in signal transduction possessing similarities to 26S
Legesse-Miller A, Elemento O, Pfau SJ, Forman JJ, Tavazoie S, Coller HA. let-7 Overex- proteasome subunits. FASEB Journal 1998;12:469–78.
pression leads to an increased fraction of cells in G2/M, direct down-regulation Shackleford TJ, Claret FX. JAB1/CSN5: a new player in cell cycle control and cancer.
of Cdc34, and stabilization of Wee1 kinase in primary fibroblasts. Journal of Cell Division 2010;5:26.
Biological Chemistry 2009;284:6605–9. Sharon M, Mao H, Boeri Erba E, Stephens E, Zheng N, Robinson CV. Symmetrical
Leppert U, Henke W, Huang X, Müller JM, Dubiel W. Post-transcriptional fine-tuning modularity of the COP9 signalosome complex suggests its multifunctionality.
of COP9 signalosome subunit biosynthesis is regulated by the c-Myc/Lin28B/let- Structure 2009;17:31–40.
7 pathway. Journal of Molecular Biology 2011;409:710–21. Shim EH, Johnson L, Noh HL, Kim YJ, Sun H, Zeiss C, et al. Expression of the F-box
Lerner M, Lundgren J, Akhoondi S, Jahn A, Ng HF, Akbari Moqadam F, et al. MiRNA-27a protein SKP2 induces hyperplasia, dysplasia, and low-grade carcinoma in the
controls FBW7/hCDC4-dependent cyclin E degradation and cell cycle progres- mouse prostate. Cancer Research 2003;63:1583–8.
sion. Cell Cycle 2011;10:2172–83. Singh A, Misra V, Thimmulappa RK, Lee H, Ames S, Hoque MO, et al. Dysfunc-
Li J, Guo Y, Liang X, Sun M, Wang G, De W, et al. MicroRNA-223 functions as an tional KEAP1-NRF2 interaction in non-small-cell lung cancer. PLOS Medicine
oncogene in human gastric cancer by targeting FBXW7/hCdc4. Journal of Cancer 2006;3:e420.
Research and Clinical Oncology 2012;138:763–74. Singhal S, Amin KM, Kruklitis R, DeLong P, Friscia ME, Litzky LA, et al. Alterations
Liakopoulos D, Doenges G, Matuschewski K, Jentsch S. A novel protein modification in cell cycle genes in early stage lung adenocarcinoma identified by expression
pathway related to the ubiquitin system. EMBO Journal 1998;17:2208–14. profiling. Cancer Biology & Therapy 2003;2:291–8.
Lin HK, Chen Z, Wang G, Nardella C, Lee SW, Chan CH, et al. Skp2 targeting Song J, Liu P, Yang Z, Li L, Su H, Lu N, et al. MiR-155 negatively regulates c-Jun
suppresses tumorigenesis by Arf-p53-independent cellular senescence. Nature expression at the post-transcriptional level in human dermal fibroblasts in vitro:
2010;464:374–9. implications in UVA irradiation-induced photoaging. Cellular Physiology and
Liu C, Powell KA, Mundt K, Wu L, Carr AM, Caspari T. Cop9/signalosome subunits Biochemistry 2012;29:331–40.
and Pcu4 regulate ribonucleotide reductase by both checkpoint-dependent and Soucy TA, Dick LR, Smith PG, Milhollen MA, Brownell JE. The NEDD8 conjuga-
-independent mechanisms. Genes and Development 2003;17:1130–40. tion pathway and its relevance in cancer biology and therapy. Genes Cancer
Liu Y, Ren S, Castellanos-Martin A, Perez-Losada J, Kwon YW, Huang Y, et al. 2010;1:708–16.
Multiple novel alternative splicing forms of FBXW7␣ have a translational mod- Takedachi A, Saijo M, Tanaka K. DDB2 complex-mediated ubiquitylation around
ulatory function and show specific alteration in human cancer. PLoS ONE DNA damage is oppositely regulated by XPC and Ku and contributes to the
2012;7:e49453. recruitment of XPA. Molecular and Cellular Biology 2010;30:2708–23.
Liu Z, Fu Q, Lv J, Wang F, Ding K. Prognostic implication of p27Kip1, Skp2 and Tang Y, Simoneau AR, Liao WX, Yi G, Hope C, Liu F, et al. WIF1, a Wnt pathway
Cks1 expression in renal cell carcinoma: a tissue microarray study. Journal of inhibitor, regulates SKP2 and c-myc expression leading to G1 arrest and growth
Experimental & Clinical Cancer Research 2008;27:51. inhibition of human invasive urinary bladder cancer cells. Molecular Cancer
Lu D, Davis MP, Abreu-Goodger C, Wang W, Campos LS, Siede J, et al. MiR-25 regulates Therapeutics 2009;8:458–68.
Wwp2 and Fbxw7 and promotes reprogramming of mouse fibroblast cells to Totary-Jain H, Sanoudou D, Dautriche CN, Schneller H, Zambrana L, Marks AR.
iPSCs. PLoS ONE 2012;7:e40938. Rapamycin resistance is linked to defective regulation of Skp2. Cancer Research
Lyapina S, Cope G, Shevchenko A, Serino G, Tsuge T, Zhou C, et al. Promotion of NEDD- 2012;72:1836–43.
CUL1 conjugate cleavage by COP9 signalosome. Science 2001;292:1382–5. Valera VA, Walter BA, Linehan WM, Merino MJ. Regulatory effects of microRNA-92
Mao JH, Perez-Losada J, Wu D, Delrosario R, Tsunematsu R, Nakayama KI, et al. (miR-92) on VHL gene expression and the hypoxic activation of miR-210 in clear
Fbxw7/Cdc4 is a p53-dependent, haploinsufficient tumour suppressor gene. cell renal cell carcinoma. Journal of Cancer 2011;2:515–26.
Nature 2004;432:775–9. van Jaarsveld MT, Helleman J, Boersma AW, van Kuijk PF, van Ijcken WF, Despierre
Misiewicz-Krzeminska I, Sarasquete ME, Quwaider D, Krzeminski P, Ticona FV, Paino E, et al. miR-141 regulates KEAP1 and modulates cisplatin sensitivity in ovarian
T, et al. Restoration of miR-214 expression reduces growth of myeloma cells cancer cells. Oncogene 2012.
through a positive regulation of P53 and inhibition of DNA replication. Haema- von Arnim AG. On again-off again: COP9 signalosome turns the key on protein
tologica 2012. degradation. Current Opinion in Plant Biology 2003;6:520–9.
Murata T, Takayama K, Katayama S, Urano T, Horie-Inoue K, Ikeda K, et al. miR- Wang B, Li W, Guo K, Xiao Y, Wang Y, Fan J. miR-181b promotes hepatic stellate cells
148a is an androgen-responsive microRNA that promotes LNCaP prostate cell proliferation by targeting p27 and is elevated in the serum of cirrhosis patients.
growth by repressing its target CAND1 expression. Prostate Cancer and Prostatic Biochemical and Biophysical Research Communications 2012a;421:4–8.
Diseases 2010;13:356–61. Wang C, Wang S, Zhao P, Wang X, Wang J, Wang Y, et al. MiR-221 promotes car-
Patil MA, Gütgemann I, Zhang J, Ho C, Cheung ST, Ginzinger D, et al. Array-based com- diac hypertrophy in vitro through the modulation of p27 expression. Journal of
parative genomic hybridization reveals recurrent chromosomal aberrations and Cellular Biochemistry 2012b;113:2040–6.
Jab1 as a potential target for 8q gain in hepatocellular carcinoma. Carcinogenesis Wei N, Chamovitz DA, Deng XW. Arabidopsis COP9 is a component of
2005;26:2050–7. a novel signaling complex mediating light control of development. Cell
1994;78:117–24.
L. Gummlich et al. / The International Journal of Biochemistry & Cell Biology 45 (2013) 1327–1337 1337

Wei N, Deng XW. Characterization and purification of the mammalian COP9 Xu Y, Sengupta T, Kukreja L, Minella AC. MicroRNA-223 regulates cyclin E activity
complex, a conserved nuclear regulator initially identified as a repressor by modulating expression of F-box and WD-40 domain protein 7. Journal of
of photomorphogenesis in higher plants. Photochemistry and Photobiology Biological Chemistry 2010;285:34439–46.
1998;68:237–41. Yamamura S, Saini S, Majid S, Hirata H, Ueno K, Deng G, et al. MicroRNA-34a
Wei N, Deng XW. The COP9 signalosome. Annual Review of Cell and Developmental modulates c-Myc transcriptional complexes to suppress malignancy in human
Biology 2003;19:261–86. prostate cancer cells. PLoS ONE 2012;7:e29722.
Wei N, Serino G, Deng XW. The COP9 signalosome: more than a protease. Trends in Yan T, Wunder JS, Gokgoz N, Gill M, Eskandarian S, Parkes RK, et al. COPS3
Biochemical Sciences 2008;33:592–600. amplification and clinical outcome in osteosarcoma. Cancer 2007;109:
Wei S, Chu PC, Chuang HC, Hung WC, Kulp SK, Chen CS. Targeting the oncogenic e3 1870–6.
ligase skp2 in prostate and breast cancer cells with a novel energy restriction- Yang ES, Burnstein KL. Vitamin D inhibits G1 to S progression in LNCaP prostate
mimetic agent. PLoS ONE 2012;7:e47298. cancer cells through p27Kip1 stabilization and Cdk2 mislocalization to the cyto-
Welcker M, Clurman BE. The SV40 large T antigen contains a decoy phosphodegron plasm. Journal of Biological Chemistry 2003;278:46862–8.
that mediates its interactions with Fbw7/hCdc4. Journal of Biological Chemistry Yang G, Ayala G, De Marzo A, Tian W, Frolov A, Wheeler TM, et al. Elevated Skp2
2005;280:7654–8. protein expression in human prostate cancer: association with loss of the cyclin-
Welcker M, Clurman BE. FBW7 ubiquitin ligase: a tumour suppressor at the dependent kinase inhibitor p27 and PTEN and with reduced recurrence-free
crossroads of cell division, growth and differentiation. Nature Reviews Cancer survival. Clinical Cancer Research 2002;8:3419–26.
2008;8:83–93. Zhang W, Kong G, Zhang J, Wang T, Ye L, Zhang X. MicroRNA-520b inhibits
Wertz IE, Kusam S, Lam C, Okamoto T, Sandoval W, Anderson DJ, et al. Sensitiv- growth of hepatoma cells by targeting MEKK2 and cyclin D1. PLoS ONE 2012;7:
ity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature e31450.
2011;471:110–4. Zhang ZW, An Y, Teng CB. The roles of miR-17-92 cluster in mammal development
White EA, Sowa ME, Tan MJ, Jeudy S, Hayes SD, Santha S, et al. Systematic identifica- and tumorigenesis. Yi Chuan 2009;31:1094–100.
tion of interactions between host cell proteins and E7 oncoproteins from diverse Zhao R, Yeung SC, Chen J, Iwakuma T, Su CH, Chen B, et al. Subunit 6 of the
human papillomaviruses. Proceedings of the National Academy of Sciences of COP9 signalosome promotes tumorigenesis in mice through stabilization of
the United States of America 2012;109:E260–7. MDM2 and is upregulated in human cancers. Journal of Clinical Investigation
Wolf DA, Zhou C, Wee S. The COP9 signalosome: an assembly and maintenance 2011;121:851–65.
platform for cullin ubiquitin ligases? Nature Cell Biology 2003;5:1029–33. Zheng J, Yang X, Harrell JM, Ryzhikov S, Shim EH, Lykke-Andersen K, et al. CAND1
Wotschofsky Z, Liep J, Meyer HA, Jung M, Wagner I, Disch AC, et al. Identifica- binds to unneddylated CUL1 and regulates the formation of SCF ubiquitin E3
tion of metastamirs as metastasis-associated MicroRNAs in clear cell renal cell ligase complex. Molecular Cell 2002;10:1519–26.
carcinomas. International Journal of Biological Sciences 2012;8:1363–74. Zheng XY, Ding W, Xie LP, Chen ZD. Correlation of Skp2 and P27kip1 pro-
Xu XM, Wang XB, Chen MM, Liu T, Li YX, Jia WH, et al. MicroRNA-19a and -19b tein expression and clinicopathological features of prostate cancer. Ai Zheng
regulate cervical carcinoma cell proliferation and invasion by targeting CUL5. 2004;23:215–8.
Cancer Letters 2012;322:148–58.

You might also like