ArticlePDF AvailableLiterature Review

A new map to understand deubiquitination: Table 1

Authors:

Abstract

Deubiquitination is a crucial mechanism in ubiquitin-mediated signalling networks. The importance of Dubs (deubiquitinating enzymes) as regulators of diverse cellular processes is becoming ever clearer as new roles are elucidated and new pathways are shown to be affected by this mechanism. Recent work, reviewed in the present paper, provides new perspective on the widening influence of Dubs and a new tool to focus studies of not only Dub interactions, but also potentially many more cellular systems.
Ubiquitin–Proteasome System, Dynamics and Targeting 21
A new map to understand deubiquitination
Elijah J. Katz, Marta Isasa and Bernat Crosas1
Institut de Biologia Molecular de Barcelona, CSIC, Barcelona Scientific Park, Baldiri i Reixac 15-21, 08028 Barcelona, Spain
Abstract
Deubiquitination is a crucial mechanism in ubiquitin-mediated signalling networks. The importance of Dubs
(deubiquitinating enzymes) as regulators of diverse cellular processes is becoming ever clearer as new roles
are elucidated and new pathways are shown to be affected by this mechanism. Recent work, reviewed in
the present paper, provides new perspective on the widening influence of Dubs and a new tool to focus
studies of not only Dub interactions, but also potentially many more cellular systems.
Introduction
The ubiquitin–proteasome system has built a complex
network of signals based on the diversity of ubiquitin
linkages and their specific recognition. One of the levels
of complexity of this fascinating regulatory system is
the reversibility of ubiquitination, which is controlled by
a variety of specialized enzymes. Factors in charge of
producing ubiquitin linkages include ubiquitin activators
(E1), ubiquitin-conjugating enzymes (E2) and ubiquitin-
ligating factors (E3 and E4) [1]. Conjugation of ubiquitin
may include the attachment of one single ubiquitin group
to protein targets, known as mono-ubiquitination, or the
formation of branches of ubiquitin polymers linked to targets,
known as polyubiquitination, which may involve all seven
lysine residues in ubiquitin (Lys6,Lys
11,Lys
27,Lys
29,Lys
33,
Lys48 and Lys63) [2]. Thus ubiquitin conjugation generates
a sophisticated code of signals in the cell, the recognition
of which is carried out by specific receptors, adaptors and
enzymes.
The enzymatic reaction that opposes ubiquitin conjugation
is deubiquitination. The human genome encodes 79 Dubs
(deubiquitinating enzymes) predicted to be active, although
most of them have not been formally characterized [3].
Catalysis of deubiquitination requires a nucleophilic attack
to the carbonyl group of the isopeptide bond established
between the C-terminal end of ubiquitin and the amino
group of the accepting lysine residue, and, in the case of
linear linkages, the attack is against a peptide bond that links
two ubiquitin groups in a head-to-tail fashion. Evolution
has found two solutions to this type of proteolytic reaction:
Key words: comparative proteomics, cytokine, deubiquitinating enzyme, signalling, ubiquitin–
proteasome system.
Abbreviations used: BRCA, breast cancer early-onset; BAP1, BRCA1-associated protein
1; CompPASS, Comparative Proteomic Analysis Software Suit; COPS, COP9 constitutive
photomorphogenic homologue subunit; CYLD, cylindromatosis; Dub, deubiquitinating enzyme;
eIF3, eukaryotic initiation factor 3; ERAD, endoplasmic-reticulum-associated degradation; GO,
Gene Ontology; HA, haemagglutinin; HCIP, high-confidence candidate-interacting protein; HEK,
human embryonic kidney; IP, immunoprecipitation; MS/MS, tandem MS; NF-κB, nuclear factor
κB; OTU, ovarian tumour protease; PHLPP, pleckstrin homology domain and leucine-rich repeat
protein phosphatase; PSMD, proteasome 26S subunit non-ATPase; STAM, signal-transducing
adaptor molecule; STAMBP, STAM-binding protein; STAMBPL1, STAMBP-like 1; UBE, ubiquitin-
conjugating enzyme; UCH, ubiquitin C-terminal hydrolase; USP, ubiquitin-specific protease; VCP,
valosin-containing protein.
1To whom correspondence should be addressed (email bernat.crosas@ibmb.csic.es).
cysteine and zinc active sites. Dubs with cysteine active sites
are more abundant and they contain a highly conserved
catalytic triad, in which an aspartic acid polarizes a histidine
residue, which deprotonates the cysteine. This mechanism
is found in UCHs (ubiquitin C-terminal hydrolases),
USPs (ubiquitin-specific proteases), OTUs (ovarian tumour
proteases) and MJDs (Machado–Josephin domains), which
represent more than 80% of human Dubs. In zinc active
sites of Dubs, similar to other metalloenzymes, a zinc atom
is attached to two histidine residues and one aspartate
residue, and one polarized water molecule co-ordinates the
fourth link to the metallic atom, ensuring reactivity. This
type of active site is found in JAMM (JAB1/MPN/MOV34
metalloenzyme) Dubs (reviewed in [3,4]).
Given the complexity of ubiquitination, Dubs have the
difficult task of discriminating their targets among an ocean
of ubiquitin–ubiquitin and protein–ubiquitin linkages in
different subcellular and functional contexts. Several layers
of specificity have been suggested to categorize Dubs: linkage
specificity (selection of ubiquitin chain topology established
by lysine usage, i.e. Lys11,Lys
48,Lys
63, etc.), relative
position of cleavage within ubiquitin chains (distal compared
with proximal cut), protein specificity (protein–ubiquitin
compared with ubiquitin–ubiquitin linkage identification),
mono-deubiquitination specificity (Dubs recognizing mono-
ubiquitinated substrates) and chain recycling (selection of
unanchored compared with anchored chains) [4]. This
classification reflects the diversity of features that define
Dubs and the number of factors selected in these enzymes.
Research on Dubs has revealed multiple regulatory roles of
deubiquitination in cellular processes; however, most Dub
enzymes have not yet been investigated.
Systematic analysis of Dub interactors
A recent study from the laboratory of Wade Harper
presented at the 2009 meeting of the INPROTEOLYS
network and published recently [5] has addressed functional
diversity of human Dubs based on their bona fide
interactors. In this work, the interactome of 73 human
Dubs was systematically analysed using proteomics. To do
so, the authors used FLAG–HA (haemagglutinin)-tagged
Biochem. Soc. Trans. (2010) 38, 21–28; doi:10.1042/BST0380021 C
The Authors Journal compilation C
2010 Biochemical Society
Biochemical Society Transactions www.biochemsoctrans.org
22 Biochemical Society Transactions (2010) Volume 38, part 1
plasmid versions of the genes expressed in HEK (human
embryonic kidney)-293 cells. They isolated Dubs by affinity
purification and analysed the protein content by LC
(liquid chromatography)–MS/MS (tandem MS) in duplicate.
To discriminate reliable interactors from non-specifically
associated proteins or simply contaminants, they developed
an unbiased approach based on a software platform called
CompPASS (Comparative Proteomic Analysis Software
Suit). For a given Dub, CompPASS assigns scores to co-
purified proteins identified within parallel proteomic analysis
in a way that proteins identified more abundantly in both
IPs (immunoprecipitations) and not common in multiple
purifications receive a higher score. HCIPs (high-confidence
candidate-interacting proteins) were defined by a novel
metric devised by the authors which takes into account
uniqueness, abundance and reproducibility of a protein
within an immunoprecipitated complex. The accuracy of
the approach was scrupulously checked using IP controls,
analysing Dubs which are part of well-established protein
complexes and by comparing the methodology with other
established techniques, as discussed below. With this tool,
they generated a list of proteins likely to be functionally
linked to each Dub and uncovered multiple interactive
networks.
Different interactive nature of Dubs
On the basis of the number and hierarchy of Dub
interactors, they found that Dubs could be assigned to
seven topological groups, each representing different types of
networking, as summarized in Table 1. Group 1 is defined
by Dubs with a low number of candidate interactors
which have not been previously described. It contains
28 members, and includes some well-characterized Dubs.
Among them, UCHL1 is involved in regulation of α2-
adrenergic receptor signalling and has been implicated in
Parkinson’s disease [6]. UCHL1 has been found to interact
with a coiled-coil domain-containing protein (CCDC14).
CYLD (cylindromatosis) is an important inhibitory regulator
of several factors involved in NF-κB (nuclear factor κB)
signal transduction [7]. An interesting interactor of CYLD
is CEP192 (centrosomal protein 192), which is involved in
mitotic spindle and centrosome assembly [8]. OTUD7B is
also involved in NF-κB-response inhibition [9], and interacts
with HIF1AN, an inhibitor of HIF1α(hypoxia-inducible
factor 1α), which is regulated by proteasome degradation
[10]. USP30 and USP33 are enzymes which have recently
been implicated in the scattering response, one of the
steps required for the invasive growth programme of
epithelial cells [11]. USP30 interacts with the MPN domain-
containing protein MPND and with USP4, which associates
with the spliceosome. OTUB1, which has been shown to
deubiquitinate and regulate oestrogen receptor αactivity
[12], interacts with two E2s [UBE (ubiquitin-conjugating
enzyme) 2N and UBE2D2]. STAMBPL1 {STAMBP [STAM
(signal-transducing adaptor molecule)-binding protein]-like
1}{or AMSH-LP [associated molecule with the SH3 (Src
homology 3) domain of STAM-like protease]}, known to
function in signal transduction, has been crystallized in
complex with Lys63-linked polyubiquitin, providing novel
structural information about this mechanism of interaction
between Dub and substrate [13]. STAMBPL1 was found to
interact with USP49, OTUB1, UBE2N and BAP1 [BRCA
(breast cancer early-onset) 1-associated protein 1]. ATXN3
has been widely studied as a cause of the neurodegenerative
Machado–Joseph disease [14] and interacts with USP13 and
OTUB2. USP16 is involved in deubiquitination of histones
in a key regulatory step of mitosis [15] and has been found to
associate with histone H2B1 and HERC2. USP28 has roles in
DNA damage response [16] and Myc stability in proliferating
tumour cells [17]. USP2 is implicated in several cancers and
in p53 regulation [18] as is USP5, also known as isopeptidase
T [19]. USP8 regulates endosomal sorting via the epidermal
growth factor receptor pathway [20]. UCHL3 has recently
been associated with metabolism and obesity control in mice
[21]. USP26 appears to be linked to infertility in males [22].
USP18 has roles in viral and bacterial pathogenesis, as well as
oncogenic transformation [23]. Other members of topology
group 1 and their interactors are shown in Table 1.
Group 2 contains Dubs that interact with or are
constituents of large macromolecular regulatory complexes.
Four of them are proteasomal Dubs, which, as expected, co-
precipitate with all proteasome subunits. They are PSMD
(proteasome 26S subunit non-ATPase) 7 (Rpn8), PSMD14
(Rpn11), UCHL5 (UCH37) and USP14 (Ubp6). UCHL5
[24] and USP14 [25] transiently associate with the regulatory
particle of the proteasome, whereas Rpn11 and Rpn8 are
integral non-ATPase subunits of the lid subcomplex [26–28].
COPS (COP9 constitutive photomorphogenic homologue
subunit) 5 and COPS6 are subunits of the COP9 signalosome
[29], which associate with the signalosome complex and with
Cullin and Kelch domain proteins. eIF3 (eukaryotic initiation
factor 3) S5 and eIF3S3 are components of the eukaryotic
initiation factor 3 complex [30]. Interestingly, the COP9
signalosome, eIF3 and the lid of the proteasome are related
complexes which show similar architecture and homologous
Dubs [31].
Group 3 consists of seven members with approx. 20–30
associated proteins which are often connected to complex
processes such as cell cycle and transcription. USP22 and
USP3 are involved in regulation of histone ubiquitination
and thus the expression of key cell-cycle components. The
former has been shown to be a component of the SAGA
(Spt–Ada–Gcn5–acetyltransferase) complex and to regulate
the expression of cell-cycle activators such as Myc [32],
whereas the latter, which is required for S-phase progression
and facilitating DNA replication [33], is found to associate
to the eIF3 complex. USP39 has been shown to be involved
in the correct expression of Aurora B kinase and other spindle
checkpoint mRNAs [34]. USP4 deubiquitinates and thus
regulates Ro52, itself a regulator of the p27 cell-cycle inhibitor
[35]. USP15 controls stability of the tumour suppressor
APC (adenomatous polyposis coli) [36]. Sowa et al. [5]
have shown that USP39, USP4 and USP15 interact with the
C
The Authors Journal compilation C
2010 Biochemical Society
Ubiquitin–Proteasome System, Dynamics and Targeting 23
Table 1 Human Dubs and their interactors
Abbreviations used in GO processes (GO-P): A, apoptosis; B, biogenesis; Cat, catabolic; Ch, chromatin; Dev, development; Dif, differentiation; DNA-D,
DNA damage; DNA-R, DNA replication; F, folding; Met, metabolic; Mit, mitosis; Mor, morphogenesis; O, other; P, proteolysis; Ph, phosphorylation;
RNA, RNA processing; ST, signal transduction; Tc, transcription; Tl, translation; Ub, ubiquitin; VT, vesicle transport. Abbreviations used in GO component
(GO-C): Cp, cytoplasm; Cs, cytoskeleton; ER, endoplasmic reticulum; G, Golgi; M, mitocondrion; N, nucleus; O, other; PM, plasma membrane; V, vesicle.
Group Dub Class Interactors GO-P GO-C
Group 1 ATXN3 MJD USP13, OTUB2, KCTD10 Ub, O N
CYLD USP SPATA2L, SPATA2, CEP192, MGEA5, CAMK2D, MYO6 Dif, Dev N, Cp
DUB3 USP LOC392197, LOC402164, CBX1, SET DNA-R, Ub, Ch N
JOSD1 MJD YOD1, CALM1, TIMM8A Dev, VT, F M
JOSD2 MJD TYSND1, KIAA0828, AHCYL1, TRAPPC2, SMARCA2 Tc, Met ER
LOC402164 USP BTF3, NACAP1, DUB3, LOC392197, ADAMTS1, LGMN, USP22 Ub, P V
OTUB1 OTU STAMBP, UBE2N, UBE2D2, MSH2, CASP14, CPNE7 Ub, A N
OTUB2 OTU GTF2I, VCPIP1 Tc, ST N
OTUD6B OTU OTUB1, ASCC3, MTDH, BXDC1 Ub, Tc N
OTUD7B OTU ACAD9, SLC9A3R2, HIF1AN, NUP155 O N
PARP11 OTU ZNF313, WDR23, LYZ Diff, Dev, Cat, Met O
STAMBPL1 JAMM/MPN USP49, OTUB1, CLINT1, UBE2N, SNRPA1, HMX3, BAP1 Ub, Tc N
UCHL1 UCH CCDC14 Dev Cs
UCHL3 UCH CLPB, USP20 - -
USP2 USP LONP1, RRP15 P M
USP5 USP USP13 Ub -
USP8 USP USP25, USP22, LOC440587 Dev Cs
USP16 USP DBT, HERC2, HIST1H2BL Ub, B, Met, VT M
USP18 USP USP41, JMJD1B, MYL6, NME1 - -
USP26 USP LOC392197, DUB3, LOC402164 Ub -
USP28 USP TP53BP1, AQR, SUMO2 N
USP29 USP USP25, CTSB A, P V, M
USP30 USP QKI, MPND, SS18L1, USP4, TIMM8A, CLPB, SF3A1 Dev, RNA N
USP33 USP ZFR, IFIT5, KRR1, PRPF38B O, RNA N
USP37 USP FBXW11, ALB Ub, A, ST, O O
USP38 USP HSPB1, HMX3, LGALS7, RPS12, RPL7 A, Tl Cp
USP48 USP LOC442227 - -
YOD1 OTU MUTED, THOC3 RNA N
Group 2 COPS5 JAMM/MPN COP9 Signalosome, Kelch domain proteins, CUL2, LRRC14, CUL4B,
WDR21A, WDR23
Ub, Mit N
COPS6 JAMM/MPN COP9 Signalosome, Kelch domain proteins, CUL4B, CUL2, LRRC14, FBXW9,
BTBD2, FBXO7, BTBD9
Ub, Mit N
EIF3S3 JAMM EIF3 complex, BAI1, MGC14327, CSNK2A2, KIAA0515, ARPC5, CSNK2B,
CSNK2A1
Tl Cp
EIF3S5 JAMM/MPN EIF3 complex, CSNK2A2, KIAA0515, CSNK2B, ASCC3, CSNK2A1 Tl Cp
PSMD7 JAMM/MPN Proteasome, KTN1 Cat Cp
PSMD14 JAMM/MPN Proteasome, FLJ20850, SUCLA2 Cat, P Cp
UCHL5 UCH Proteasome, NFRKB, TFPT, TXNL1, CCDC95, PTPN2 Ub, Cat Cp
USP14 USP Proteasome, TXNL1, RGPD5, SRPRB Ub, Cat Cp
Group 3 JOSD3 MJD Transcriptional complexes, POLR1E, CENPB, RRP15, POLR1C, PPAN,
LOC440587
Ts N
USP3 USP EIF3 complex, WDTC1, RIMBP2, LRP1, NXN, USP48, GNAL, GNA13, CBR3,
IFRG15, PKLR, KIAA0515
Tl Cp
USP4 USP Spliceosome complex, EDC3, AKAP7, PRPF3, TUT1, DCP1B, BCDIN3, ADSL,
USP32, EIF4E2
RNA N
USP15 USP Spliceosome complex, LRRC15, RNF40, MYH4, SELENBP1, FABP4, VSIG8,
MYH2, TUT1, PRPF3, ADSL
RNA N
USP22 USP SAGA complex, ENY2, TADA1L, LOC254571, ATXN7L(3,2), MGC21874, KIF7,
FAM48A, LOC552889, USP27X
Tc N
C
The Authors Journal compilation C
2010 Biochemical Society
24 Biochemical Society Transactions (2010) Volume 38, part 1
Table 1 Continued
Group Dub Class Interactors GO-P GO-C
USP39 USP Spliceosome complex, RG9MTD1, PA2G4, ZRANB2, RY1, PRPF(4B, 3, 4),
TSR2, KIAA0409, C17orf79
RNA N
ZRANB1 OTU Phosphatase scaffolding complexes, CTTNBP2NL, HECTD1, MAP4K4, GCC2,
KEAP1, PGAM5, CACYBP
Ub, O Cp
Group 4 OTUD1 OTU LOC653852, FLNC, RAD23A, RAD23B, FLNB, FLNA, KEAP1 DNA-D,ST, Dev, B N, Cs
STAMBP JAMM/MPN VPS24, AFP, PIK3C2A, OTUB1, CLINT1, GRB2, CLTA ST, VT V, O
TL132L USP USP5, LOC220594, CYLD, USP13, USP32, RNH1, SEH1L, OTUB2, TGM3 Ub Cs
USP10 USP CUGBP1, EIF4G1, EIF4G3, G3BP2, G3BP1, NOLA1 ST, Tl, RNA Cp
Group 5 BAP1 UCH OGT, HCFC1, RBBP7, HAT1, FOXK1, UBE20, FOXK2, ASXL2, ANKRD17,
EIF4EBP3, ASXL1, IPO4, CBX3
Tc N
BRCC3 JAMM/MPN KIAA0157, HSPC142, BRE, CCDC98, UIMC1, EXOC8, SUPT16H, E2F5, DDOST,
MCM2, CAND1, SHMT2
Tc N
OTUD4 OTU GLA, GALK1, DSG1, PARP11, MYCBP, NMD3, MOG, GPHN, BAG5, DNAJB1,
FLNC, TUBA1A
OCs
USP1 USP PKP1, DSC1, DSG1, JUP, USP3, CALML3, TAGLN2, HSPB1, CALML5, WDR48,
PHLPP, MYH9, KPNA1
Dev Cs
USP13 USP DLST, OGDH, SMC1A, SMC3, UBL4A, UFD1L, DIABLO, KCTD3, KCTD10, ITCH,
NPLOC4, UBXD8, CACYBP
Ub N
USP21 USP RBM8A, UPF3B, MARK3, UCHL1, MARK(1,2), KIAA1553, UTRN, FUCA1,
MARK4, PRKCI, USP20, USP48
Ph, Dev N, Cp
USP25 USP PIP, LYZ, LOC124220, WRNIP1, USP28, KCTD13, ANXA1, KLHL9, KCTD10,
BTBD9, MYO6, NEDD8
-N
USP32 USP TUBA1A, CDC2, YWHAB, USP6, VPS35, LOC51035, MRPL39, ABCD3, USP11,
TRIP13, TXNDC4, SMC1A
ON
USP36 USP WDR36, WDR3, UTP18, PWP2, TBL3, DHX33, NUDCD1, MPND, STK25,
DDX41, GNL2, CHD4, TMEM104
ST N
USP45 USP SF3A2, RBMX, POLR2G, MYH10, CORO1C, MRLC2, RTCD1, SRBD1, MYH9,
TMOD3, PIK3CG, GLTSCR2
RNA N
USP50 USP ABCE1, SLC1A5, DYNC2H1, LUC7L, FLJ20294, IGF2R, KEAP1, AMOT, CKAP4,
AFG3L2, VCP complex
OM,N
USP52 USP VBP1, TCEB2, TCEB1, PFDN2, PAN3, USP5, PLEC1, NUP93, MRPL39, ARCN1,
CBWD2, CCT7
FN
USPL1 USP PGD, PKLR, ANKFY1, KIAA0947, ELL, KIF5B, IGHA2, CKB, ANXA1, PIP O Cp, Cs, O
VCPIP1 OTU NSFL1C, UFD1L, NPLOC4, VCP, UBXD4, ABCC12, LOC137886,
KFZp313A2432, UBXD8, HUWE1
Ub N
Group 6 OTUD5 OTU LONRF2, GPX4, LANCL2, CTPS2, GYS1, VARS, FLNA, CACYBP, SET, GRB2,
USP11, TP53, PKLR
Dev N
TNFAIP3 OTU KIF11, NDUFS1, GLDC, FBXO3, CNKSR2, TBK1, YWHAH, RNH1, YWHAB,
LRRC47, ALDH9A1, PPP2R1B
-Cp
USP12 USP USP39, WDR20, DMWD, PHLPP, PHLPPL, UCHL5, WDR48, RAD51AP1, CYLD,
PAIP1, NUP160, MMP2
RNA N
USP42 USP PLRG1, TEX10, C14orf169, AMOT, USP32, AHCYL1, DIMT1L, USP20, FECH,
MRPS14, CA2, MRPS31
-N
USP43 USP MAGI3, WDR3, YWHAH, PDCD2, YWHAB, VPS35, FN3KRP, PSME3, YWHAE A, ST Cp
USP44 USP CETN2, MRPL40, SARS2, POLR2G, MRPL53, MRPL23, TCOF1, KRR1, TBL2,
MRPS21
Tl, O M, N
USP46 USP USP12, WDR20, DMWD, PHLPP, IQGAP1, PHLPPL, EIF2AK4, PPP1R9B,
WDR48, RAD51AP1, PJA2, USP52
Ub, Ph Cp
USP53 USP ARG1, OTUD4, CAT, CSTA, BLMH, CASP14, HCFC1, AZGP1, TGM3, RPL26,
RAD50, DSG1, KEAP1
Mit Cp
Group 7 USP7 USP MCM6, NUP98, MCM4, CRKL, MCM5, RAE1, C10orf119, USP14, BRCC3,
KIAA0157, FBXO38, USP19, BRE
Ub, Tc N
C
The Authors Journal compilation C
2010 Biochemical Society
Ubiquitin–Proteasome System, Dynamics and Targeting 25
Table 1 Continued
Group Dub Class Interactors GO-P GO-C
USP11 USP SIPA1L1, ZNF24, USP4, MRE11A, USP7, RAD50, TP53, PKN2, TCEAL(1,4),
TRIP12, MPHOSPH9, OTUD1
Tc N
USP19 USP CSE1L, CDC2, HMOX2, TNPO3, CAMK2G, LAT, DMWD, KIAA1787, HERC2,
ACADSB, PARK7, UNC45A
VT N
USP20 USP PSMD6, EIF3S6, PSMD7, EIF3S8, PSMD11, PSMD12, VAPA, PLEKHA7,
RAD17, SYTL4, N4BP3, UCHL3
ON,Cp
USP49 USP PDCD2, LRPPRC, Centrin proteins, SAPS domain proteins, USP44, F7, FKBP5,
SHCBP1, PPP5C, ANKRD28
VT N
spliceosome complex. JOSD3 (Josephin domain-containing
3) is implicated in carcinogensis and RNA polymerase I
transcription of rRNA [37,38], and binds to transcriptional
complexes.
The four members of Group 4 are so-called ‘distributive’
Dubs with a low number of interactors (six to eight) that
may function in several distinct pathways. USP10 interacts
with G3BP2 [GTPase-activating protein (Src homology
3 domain)-binding protein 2], an activator of the Ras
signal transduction pathway, as well as with eukaryotic
initiation factors. It is also involved in the regulation
of androgen receptor function [39]. STAMBP has known
regulatory relationships with the calcium-sensing receptor
pathway and endosomal sorting of epidermal growth factor
[40]. CompPASS analysis confirmed interaction with these
pathways as well as linking STAMBP to the Ras pathway.
Group 5 Dubs have approx. 10–20 interactors, some
exhibiting likely interconnectivity and others which appear
to be unrelated. Specific functions of many members of
this group are as yet unstudied. USP36 is involved in
regulation of nucleolus structure and function, affecting
the synthesis of rRNA [41]. BAP1 is a putative tumour
suppressor that interacts with the BRCA1 protein and has
a role in DNA-damage repair [42]. USP1 deubiquitinates
mono-ubiquitinated FANCD2 (Fanconi’s anaemia comple-
mentation group D2), a component of the Fanconi’s anemia
DNA-repair pathway [43]. BRCC6 is a BRCA1/BRCA2-
containing complex subunit with a role in ERK (extracellular-
signal-regulated kinase) signal transduction [44]. USP25
is a Dub with multiple splice variants whose activity
has been shown to be alternatively regulated by mono-
ubiquitination and SUMOylation [45]. Sowa et al. [5]
suggest an interaction of USP25 with a BTB/POZ domain-
containing protein NEDD8 (neural-precursor-cell-expressed
developmentally down-regulated 8), another ubiquitin-
like signalling molecule. VCPIP1 [VCP (valosin-containing
protein)/p97–p47 complex-interacting protein 1] is involved
in p97–p47-mediated membrane fusion and assembly of
Golgi and endoplasmic reticulum structures [46].
The Group 6 Dubs are characterized by having a large
number of interactors with highly distributive functional
implications. TNFAIP3 (tumour necrosis factor α-induced
protein 3) plays an important role in the inhibition of NF-κB
responses [47]. USP44 is part of a mechanism which controls
the initiation of anaphase during mitosis [48]. OTUD5 is an
inhibitor of innate immune response [49].
Group 7 contains some Dubs that are known to have
roles in diverse processes. Their interaction topologies are
characterized by a large number of interaction candidates,
where a small subset may have apparent interconnectivity,
but the majority appear to be unrelated or distributive. USP7
has roles in regulation on the p53 tumour suppressor and
in the life cycles of several human herpesviruses [50,51].
USP11 also acts in the p53 pathway, as well as the BRCA2
DNA-damage repair pathway and participates in the life
cycle of human papillomavirus 16 [52,53,54]. USP20 has been
implicated in endocytic sorting and β2-adrenergic receptor-
mediated signalling [55].
Overall, human Dubs show a wide repertoire of inter-
actors. A challenging aspect is the high number of previously
unknown interactions, a selection of which is presented in
Table 1.
Dub interactome validation
The efficacy of the CompPASS system at accurately
identifying reproducible protein–protein interactions was
confirmed both experimentally and by comparison with
published literature. IPs of 25 HA-tagged interactors that
were subjected to IP–MS/MS confirmed 83% of CompPASS
predicted interactions. Co-immunoprecipitation experiments
using Myc-tagged interactors and Dubs validated 14 of
29 predicted interactions by immunoblotting. Endogenous
co-immunoprecipitation detected three of six predicted
interactions. Overall, 68% of interactions tested were
confirmed independently. Study of the literature revealed
332 interactions involving 51 different Dubs. CompPASS
prediction shared 71% identity with published interactions
identified using endogenous co-IP or co-purification. This
value was 36% when compared with interactions charac-
terized by overexpression co-IP and just 4.6% compared
with interactions characterized by yeast two-hybrid systems
alone. Finally, when 11 Dubs from the set were expressed
in HCT116 cells and subjected to IP–MS/MS, CompPASS
C
The Authors Journal compilation C
2010 Biochemical Society
26 Biochemical Society Transactions (2010) Volume 38, part 1
analysis identified 63% of the same HCIPs from previous
HEK-293T cell samples. With this kind of accuracy in the
identification of interacting partners, CompPASS can help to
point out potentially fruitful avenues for future research into
the breadth and depth of the Dub interactome.
Following validation procedures, robust interactions can
be established, such as the involvement of USP13 in ERAD
(endoplasmic-reticulum-associated degradation). USP13 was
found to interact with the AAA (ATPase associated with
various cellular activities) VCP/p97 complex and with the
VCP-associated proteins UFD1 (ubiquitin fusion degrada-
tion 1), NPL4 (nuclear protein localization 4) and UBDX8.
The authors performed functional studies and showed that
depletion of USP13 increases the levels of the ERAD
substrate TCRα(T-cell receptor α)–GFP (green fluorescent
protein), suggesting an relevant role of this Dub in the
pathway.
The authors also found interesting interconnectivity
of related Dubs USP1, USP12 and USP46 with two
phosphatases involved in Akt dephosphorylation [PHLPP
(pleckstrin homology domain and leucine-rich repeat protein
phosphatase) and PHLPPL (PHLPP-like)] [56] and three
WD40-containing proteins [WDR48/UAF1, WDR20 and
DMWD (dystrophia myotonica, WD40 repeat-containing)].
The capacity of WDR48/UAF1 to associate with USP1,
USP12 and USP46 has been reported [57,58], but WDR20
and DMWD are uncharacterized. It has been shown recently
that PHLPP is polyubiquitinated and degraded in a β-TrCP
(β-transducin repeat-containing protein)-dependent manner
and that Akt regulates this process [59], suggesting that a
cognate Dub might be involved in PHLPP stability.
Additional Dub-containing networks described in the
present paper are related to RNA processing, DNA-damage
response and ribosome autophagy. For instance, USP39,
USP15 and USP4 were found in U5/U6-snRNP (small
nuclear riboprotein) and in Lsm mRNA-binding complexes.
Another interesting finding is that USP11, a p53 regulator,
was verified to interact with USP7 and USP4, which have
been implicated in the p53 and p27 pathways respectively
[29,44,46]. Determination of relationships could lead to
greater understanding of the role of ubiquitin signalling in the
wider context of global cell-cycle regulation. Furthermore,
USP11 and USP7 share interactors with roles in cell-cycle
regulation, including RAE1 (RNA export 1) [60] and BUB3
(budding uninhibited by benzimidazoles 3) [61].
A general feature is that multiple Dubs tend to
associate with a given network or pathway, suggesting that
deubiquitination may regulate several steps or levels of the
pathway.
An exportable workflow
The present paper defines the workflow for a new generation
of systematic proteomic studies. The first step is processing
raw data from proteomic analysis. The high and ever-
increasing sensitivity of protein MS requires appropriate
bioinformatics implementation. In this regard, CompPASS
defines a successful rationale to process raw data. The
second step is to provide a cellular and functional context
to new interactions. By categorizing networks according
to GO (Gene Ontology) and subcellular localization, a
functional landscape is plotted. The third and very important
step is validating novel cellular networks. The relevance
of comparative proteomics analysis is that it outlines
putative interactomes which define multiple novel functions
for uncharacterized proteins. These putative interactomes
must be validated experimentally and this is what Sowa
et al. [5] have done. By reciprocal tagging of interactor
candidates, and by using other techniques not based on
MS analysis, they have defined robust interactions and
validated novel actors. As a sample of how powerful the
approach could be, by validating interactions, they propose
novel roles for Dubs in Akt regulation, RNA processing,
DNA-damage response, ribophagy and ERAD. Overall, this
approach appears to be a tool exportable to almost any
group of proteins of interest. Currently, CompPASS is a
resource available online to any researcher (http://pathology.
hms.harvard.edu/labs/harper/CompPASS.html). The me-
thod cannot follow up those interactions that, although
being functionally relevant, are not reproduced in IPs, but
this caveat is compensated for by the fact that it rationally
substantiates the study of hundreds, if not more, of novel
uncharacterized proteins.
Funding
This work was supported by the Spanish Government Ministerio de
Educaci ´on y Ciencia [grant number BFU2006-02928]
References
1 Finley, D. (2009) Recognition and processing of ubiquitin-protein
conjugates by the proteasome. Annu. Rev. Biochem. 78, 477–513
2 Xu, P., Duong, D.M., Seyfried, N.T., Cheng, D., Xie, Y., Robert, J., Rush, J.,
Hochstrasser, M., Finley, D. and Peng, J. (2009) Quantitative proteomics
reveals the function of unconventional ubiquitin chains in proteasomal
degradation. Cell 137, 133–145
3 Nijman, S.M., Luna-Vargas, M.P., Velds, A., Brummelkamp, T.R., Dirac,
A.M., Sixma, T.K. and Bernards, R. (2005) A genomic and functional
inventory of deubiquitinating enzymes. Cell 123, 773–786
4 Komander, D., Clague, M.J. and Urb ´e, S. (2009) Breaking the chains:
structure and function of the deubiquitinases. Nat. Rev. Mol. Cell Biol.
10, 550–563
5 Sowa, M.E., Bennett, E.J., Gygi, S.P. and Harper, J.W. (2009) Defining the
human deubiquitinating enzyme interaction landscape. Cell 138,
389–403
6 Weber, B., Schaper, C., Wang, Y., Scholz, J. and Bein, B. (2009)
Interaction of the ubiquitin carboxyl terminal esterase L1 with
α2-adrenergic receptors inhibits agonist-mediated p44/42 MAP kinase
activation. Cell. Signalling 21, 1513–1521
7 Trompouki, E., Hatzivassiliou, E., Tsichritzis, T., Farmer, H., Ashworth, A.
and Mosialos, G. (2003) CYLD is a deubiquitinating enzyme that
negatively regulates NF-κB activation by TNFR family members. Nature
424, 793–796
8 Gomez-Ferreria, M.A., Rath, U., Buster, D.W., Chanda, S.K., Caldwell, J.S.,
Rines, D.R. and Sharp, D.J. (2007) Human Cep192 is required
for mitotic centrosome and spindle assembly. Curr. Biol. 17,
1960–1966
9 Enesa, K., Zakkar, M., Chaudhury, H., Luong le, A., Rawlinson, L., Mason,
J.C., Haskard, D.O., Dean, J.L. and Evans, P.C. (2008) NF-κB suppression
by the deubiquitinating enzyme Cezanne: a novel negative feedback
loop in pro-inflammatory signaling. J. Biol. Chem. 283, 7036–7045
C
The Authors Journal compilation C
2010 Biochemical Society
Ubiquitin–Proteasome System, Dynamics and Targeting 27
10 Jariel-Encontre, I., Bossis, G. and Piechaczyk, M. (2008)
Ubiquitin-independent degradation of proteins by the proteasome.
Biochem. Biophys. Acta 1786, 153–177
11 Buus, R., Faronato, M., Hammond, D.E., Urb ´e, S. and Clague, M.J. (2009)
Deubiquitinase activities required for hepatocyte growth factor-induced
scattering of epithelial cells. Curr. Biol. 19, 1463–1466
12 Stanisic, V., Malovannaya, A., Qin, J., Lonard, D.M. and O’Malley, B.W.
(2009) OTU domain-containing ubiquitin aldehyde-binding protein 1
(OTUB1) deubiquitinates estrogen receptor (ER) αand affects ERα
transcriptional activity. J. Biol. Chem. 284, 16135–16145
13 Sato, Y., Yoshikawa, A., Yamagata, A., Mimura, H., Yamashita, M.,
Ookata, K., Nureki, O., Iwai, K., Komada, M. and Fukai, S. (2008)
Structural basis for specific cleavage of Lys63-linked polyubiquitin chains.
Nature 455, 358–362
14 Boeddrich, A., Gaumer, S., Haacke, A., Tzvetkov, N., Albrecht, M., Evert,
B.O., M ¨uller, E.C., Lurz, R., Breuer, P., Schugardt, N. et al. (2006) An
arginine/lysine-rich motif is crucial for VCP/p97-mediated modulation of
ataxin-3 fibrillogenesis. EMBO J. 25, 1547–1558
15 Joo, H.Y., Zhai, L., Yang, C., Nie, S., Erdjument-Bromage, H., Tempst, P.,
Chang, C. and Wang, H. (2007) Regulation of cell cycle progression
and gene expression by H2A deubiquitination. Nature 449,
1068–1072
16 Zhang, D., Zaugg, K., Mak, T.W. and Elledge, S.J. (2006) A role for the
deubiquitinating enzyme USP28 in control of the DNA-damage response.
Cell 126, 529–542
17 Popov, N., Wanzel, M., Madiredjo, M., Zhang, D., Beijersbergen, R.,
Bernards, R., Moll, R., Elledge, S.J. and Eilers, M. (2007) The
ubiquitin-specific protease USP28 is required for MYC stability. Nat. Cell
Biol. 9, 765–774
18 Stevenson, L.F., Sparks, A., Allende-Vega, N., Xirodimas, D.P., Lane, D.P.
and Saville, M.K. (2007) The deubiquitinating enzyme USP2a regulates
the p53 pathway by targeting Mdm2. EMBO J. 26, 976–986
19 Dayal, S., Sparks, A., Jacob, J., Allende-Vega, N., Lane, D.P. and Saville,
M.K. (2009) Suppression of the deubiquitinating enzyme USP5 causes
the accumulation of unanchored polyubiquitin and the activation of p53.
J. Biol. Chem. 284, 5030–5041
20 Alwan, H.A. and van Leeuwen, J.E. (2007) UBPY-mediated epidermal
growth factor receptor (EGFR) de-ubiquitination promotes EGFR
degradation. J. Biol. Chem. 282, 1658–1669
21 Setsuie, R., Suzuki, M., Kabuta, T., Fujita, H., Miura, S., Ichihara, N.,
Yamada, D., Wang, Y.L., Ezaki, O., Suzuki, Y. and Wada, K. (2009)
Ubiquitin C-terminal hydrolase-L3-knockout mice are resistant to
diet-induced obesity and show increased activation of AMP-activated
protein kinase in skeletal muscle. FASEB J. 23, 4148–4157
22 Ribarski, I., Lehavi, O., Yogev, L., Hauser, R., Bar-Shira Maymon, B.,
Botchan, A., Paz, G., Yavetz, H. and Kleiman, S.E. (2009) USP26
gene variations in fertile and infertile men. Hum. Reprod. 24,
477–484
23 Yan, M., Luo, J.K., Ritchie, K.J., Sakai, I., Takeuchi, K., Ren, R. and Zhang,
D.E. (2007) Ubp43 regulates BCR-ABL leukemogenesis via the type 1
interferon receptor signaling. Blood 110, 305–312
24 Qiu, X.B., Ouyang, S.Y., Li, C.J., Miao, S., Wang, L. and Goldberg, A.L.
(2006) hRpn13/ADRM1/GP110 is a novel proteasome subunit that
binds the deubiquitinating enzyme, UCH37. EMBO J. 25, 5742–5753
25 Borodovsky, A., Kessler, B.M., Casagrande, R., Overkleeft, H.S., Wilkinson,
K.D. and Ploegh, H.L. (2001) A novel active site-directed probe specific
for deubiquitylating enzymes reveals proteasome association of USP14.
EMBO J. 20, 5187–5196
26 Tsurumi, C., DeMartino, G.N., Slaughter, C.A., Shimbara, N. and Tanaka, K.
(1995) cDNA cloning of p40, a regulatory subunit of the human 26S
proteasome, and a homolog of the Mov-34 gene product. Biochem.
Biophys. Res. Commun. 210, 600–608
27 Spataro, V., Toda, T., Craig, R., Seeger, M., Dubiel, W., Harris, A.L. and
Norbury, C. (1997) Resistance to diverse drugs and ultraviolet light
conferred by overexpression of a novel human 26 S proteasome subunit.
J. Biol. Chem. 272, 30470–30475
28 Sharon, M., Taverner, T., Ambroggio, X.I., Deshaies, R.J. and Robinson,
C.V. (2006) Structural organization of the 19S proteasome lid: insights
from MS of intact complexes. PLoS Biol. 4, e267
29 Schwechheimer, C. (2004) The COP9 signalosome (CSN): an evolutionary
conserved proteolysis regulator in eukaryotic development. Biochem.
Biophys. Acta 1695, 45–54
30 Masutani, M., Sonenberg, N., Yokoyama, S. and Imataka, H. (2007)
Reconstitution reveals the functional core of mammalian eIF3. EMBO
J. 26, 3373–3383
31 Scheel, H. and Hofmann, K. (2005) Prediction of a common structural
scaffold for proteasome lid, COP9-signalosome and eIF3 complexes. BMC
Bioinformatics 6,71
32 Zhang, X.Y., Varthi, M., Sykes, S.M., Phillips, C., Warzecha, C., Zhu, W.,
Wyce, A., Thorne, A.W., Berger, S.L. and McMahon, S.B. (2008) The
putative cancer stem cell marker USP22 is a subunit of the human SAGA
complex required for activated transcription and cell-cycle progression.
Mol. Cell 29, 102–111
33 Nicassio, F., Corrado, N., Vissers, J.H., Areces, L.B., Bergink, S., Marteijn,
J.A., Geverts, B., Houtsmuller, A.B., Vermeulen, W., Di Fiore, P.P. and
Citterio, E. (2007) Human USP3 is a chromatin modifier required for S
phase progression and genome stability. Curr. Biol. 17, 1972–1977
34 van Leuken, R.J., Luna-Vargas, M.P., Sixma, T.K., Wolthuis, R.M. and
Medema, R.H. (2008) Usp39 is essential for mitotic spindle checkpoint
integrity and controls mRNA-levels of aurora B. Cell Cycle 7, 2710–2719
35 Wada, K. and Kamitani, T. (2006) UnpEL/Usp4 is ubiquitinated by Ro52
and deubiquitinated by itself. Biochem. Biophys. Res. Commun. 342,
253–258
36 Huang, X., Langelotz, C., Hetfeld-Pechoc, B.K., Schwenk, W. and Dubiel,
W. (2009) The COP9 signalosome mediates β-catenin degradation by
deneddylation and blocks adenomatous polyposis coli destruction via
USP15. J. Mol. Biol. 391, 691–702
37 Wang, L., Bhattacharyya, N., Chelsea, D.M., Escobar, P.F. and Banerjee, S.
(2004) A novel nuclear protein, MGC5306 interacts with DNA
polymerase βand has a potential role in cellular phenotype. Cancer Res.
64, 7673–7677
38 Gorski, J.J., Pathak, S., Panov, K., Kasciukovic, T., Panova, T., Russell, J. and
Zomerdijk, J.C. (2007) A novel TBP-associated factor of SL1 functions in
RNA polymerase I transcription. EMBO J. 26, 1560–1568
39 Faus, H., Meyer, H.A., Huber, M., Bahr, I. and Haendler, B. (2005) The
ubiquitin-specific protease USP10 modulates androgen receptor function.
Mol. Cell. Endocrinol. 245, 138–146
40 Herrera-Vigenor, F., Hern ´andez-Garc´ıa, R., Valadez-S ´anchez, M.,
azquez-Prado, J. and Reyes-Cruz, G. (2006) AMSH regulates
calcium-sensing receptor signaling through direct interactions. Biochem.
Biophys. Res. Commun. 347, 924–930
41 Endo, A., Matsumoto, M., Inada, T., Yamamoto, A., Nakayama, K.I.,
Kitamura, N. and Komada, M. (2009) Nucleolar structure and function
are regulated by the deubiquitylating enzyme USP36. J. Cell Sci. 122,
678–686
42 Jensen, D.E., Proctor, M., Marquis, S.T., Gardner, H.P., Ha, S.I., Chodosh,
L.A., Ishov, A.M., Tommerup, N., Vissing, H., Sekido, Y. et al. (1998)
BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger
and enhances BRCA1-mediated cell growth suppression. Oncogene 16,
1097–1112
43 Nijman, S.M., Huang, T.T., Dirac, A.M., Brummelkamp, T.R., Kerkhoven,
R.M., D’Andrea, A.D. and Bernards, R. (2005) The deubiquitinating
enzyme USP1 regulates the Fanconi anemia pathway. Mol. Cell 17,
331–339
44 Boudreau, H.E., Broustas, C.G., Gokhale, P.C., Kumar, D., Mewani, R.R.,
Rone, J.D., Haddad, B.R. and Kasid, U. (2007) Expression of BRCC3, a
novel cell cycle regulated molecule, is associated with increased
phospho-ERK and cell proliferation. Int. J. Mol. Med. 19, 29–39
45 Denuc, A., Bosch-Comas, A., Gonz `alez-Duarte, R. and Marfany, G. (2009)
The UBA-UIM domains of the USP25 regulate the enzyme ubiquitination
state and modulate substrate recognition. PLoS ONE 4, e5571
46 Uchiyama, K., Jokitalo, E., Kano, F., Murata, M., Zhang, X., Canas, B.,
Newman, R., Rabouille, C., Pappin, D., Freemont, P. and Kondo, H.
(2002) VCIP135, a novel essential factor for p97/p47-mediated
membrane fusion, is required for Golgi and ER assembly in vivo.J.Cell
Biol. 159, 855–866
47 Wertz, I.E., O’Rourke, K.M., Zhou, H., Eby, M., Aravind, L., Seshagiri, S.,
Wu, P., Wiesmann, C., Baker, R., Boone, D.L. et al. (2004)
De-ubiquitination and ubiquitin ligase domains of A20 downregulate
NF-κB signalling. Nature 430, 694–699
48 Stegmeier, F., Rape, M., Draviam, V.M., Nalepa, G., Sowa, M.E., Ang, X.L.,
McDonald, 3rd, E.R., Li, M.Z., Hannon, G.J., Sorger, P.K. et al. (2007)
Anaphase initiation is regulated by antagonistic ubiquitination and
deubiquitination activities. Nature 446, 876–881
49 Kayagaki, N., Phung, Q., Chan, S., Chaudhari, R., Quan, C., O’Rourke, K.M.,
Eby, M., Pietras, E., Cheng, G., Bazan, J.F. et al. (2007) DUBA: a
deubiquitinase that regulates type I interferon production. Science 318,
1628–1632
50 Li, M., Chen, D., Shiloh, A., Luo, J., Nikolaev, A.Y., Qin, J. and Gu, W.
(2002) Deubiquitination of p53 by HAUSP is an important pathway for
p53 stabilization. Nature 416, 648–653
C
The Authors Journal compilation C
2010 Biochemical Society
28 Biochemical Society Transactions (2010) Volume 38, part 1
51 Antrobus, R. and Boutell, C. (2008) Identification of a novel higher
molecular weight isoform of USP7/HAUSP that interacts with the Herpes
simplex virus type-1 immediate early protein ICP0. Virus Res. 137, 64–71
52 Yamaguchi, T., Kimura, J., Miki, Y. and Yoshida, K. (2007) The
deubiquitinating enzyme USP11 controls an IκBkinaseα(IKKα)-p53
signaling pathway in response to tumor necrosis factor α(TNFα). J. Biol.
Chem. 282, 33943–33948
53 Schoenfeld, A.R., Apgar, S., Dolios, G., Wang, R. and Aaronson, S.A.
(2004) BRCA2 is ubiquitinated in vivo and interacts with USP11, a
deubiquitinating enzyme that exhibits prosurvival function in the cellular
response to DNA damage. Mol. Cell. Biol. 24, 7444–7455
54 Lin, C.H., Chang, H.S. and Yu, W.C. (2008) USP11 stabilizes HPV-16E7 and
further modulates the E7 biological activity. J. Biol. Chem. 283,
15681–15688
55 Berthouze, M., Venkataramanan, V., Li, Y. and Shenoy, S.K. (2009) The
deubiquitinases USP33 and USP20 coordinate β2adrenergic receptor
recycling and resensitization, EMBO J. 28, 1684–1696
56 Gao, T., Furnari, F. and Newton, A.C. (2005) PHLPP: a phosphatase that
directly dephosphorylates Akt, promotes apoptosis, and suppresses
tumor growth. Mol. Cell 18, 13–24
57 Cohn, M.A., Kowal, P., Yang, K., Haas, W., Huang, T.T., Gygi, S.P. and
D’Andrea, A.D. (2007) A UAF1-containing multisubunit protein complex
regulates the Fanconi anemia pathway. Mol. Cell 28, 786–797
58 Cohn, M.A., Kee, Y., Haas, W., Gygi, S.P. and D’Andrea, A.D. (2009) UAF1
is a subunit of multiple deubiquitinating enzyme complexes. J. Biol.
Chem. 284, 5343–5351
59 Li, X., Liu, J. and Gao, T. (2009) β-TrCP-mediated ubiquitination and
degradation of PHLPP1 is negatively regulated by Akt. Mol. Cell. Biol. 29,
6192–6205
60 Wong, R.W., Blobel, G. and Coutavas, E. (2006) Rae1 interaction with
NuMA is required for bipolar spindle formation. Proc. Natl. Acad. Sci.
U.S.A. 103, 19783–19787
61 Yoon, Y.M., Baek, K.H., Jeong, S.J., Shin, H.J., Ha, G.H., Jeon, A.H., Hwang,
S.G., Chun, J.S. and Lee, C.W. (2004) WD repeat-containing mitotic
checkpoint proteins act as transcriptional repressors during interphase.
FEBS Lett. 575, 23–29
Received 23 October 2009
doi:10.1042/BST0380021
C
The Authors Journal compilation C
2010 Biochemical Society
... While there is a complex ubiquitination system for ubiquitinating proteins and target them for modification or degradation, there also correspondingly exists a deubiquitination program [19]. It is the process of cleaving ubiquitin molecules from ubiquitin conjugated protein substrates by deubiquitinating enzymes (DUBs), which can rescue proteins from degradative pathways or lead to reversion of ubiquitin signaling [20][21][22]. Over one hundred genes potentially coding for DUBs are identified in human genome and can be classified into five different varieties including JAB1/MPN/Mov34 (JAMM), ubiquitin C-terminal hydrolase (UCHs), ubiquitin-specific protease (USPs), ovarian tumor domain (OTUDs) and Josephin. ...
... Over one hundred genes potentially coding for DUBs are identified in human genome and can be classified into five different varieties including JAB1/MPN/Mov34 (JAMM), ubiquitin C-terminal hydrolase (UCHs), ubiquitin-specific protease (USPs), ovarian tumor domain (OTUDs) and Josephin. JAMM belongs to the superfamily of zinc-dependent metalloproteases, and other DUBs are classical papain-like cysteine proteases [20,21]. Cezanne (OTUD7B), a member of OTUDs family, shows a preference for K11-uniquitin linkages [23]. ...
Article
Background: Degradation of insulin-like growth factor 1 receptor (IGF-1R) is mediated by internalization and endocytosis, for which ubiquitin-proteasome pathways play as a regulatory system. Cezanne expression is positively associated with IGF-1R expression. High Cezanne expression correlates with poor patient survival in NSCLC, yet the underlying mechanisms are not well defined. Methods: Co-Immunoprecipitation assay was performed to investigate the interactions between Cezanne and IGF-1R. A xenograft model was established to assess the efficacy of Cezanne on cancer progression in vivo. Cezanne overexpressing and Cezanne knockdown NSCLC cell lines were generated using lentiviral vectors. The effects of Cezanne and IGF-1R on cell proliferation of non-small-cell lung cancer were evaluated via Sulforhodamine B assay and colony formation assays. Results: Here, through co-Immunoprecipitation assay, we find Cezanne interacts with IGF-1R in tumor cells. Depletion of Cezanne promotes the ubiquitination and degradation of IGF-1R. Congruently, Cezanne regulates the protein level of IGF-1R and downstream AKT signaling pathway. Cezanne promotes proliferation of tumor cells in vitro and in vivo. In line with the change of IGF-1R downstream signaling pathway, IGF-1-induced growth signals recover cell proliferation of tumor cells with Cezanne knockdown. Conclusion: Mechanistically, Cezanne directly targets IGF-1R by deubiquitination and stabilization. This leads to AKT activation, which bolsters tumor cell growth in vitro and in vivo. These findings reveal Cezanne as a regulator of tumor cell proliferation via IGF-1R signaling pathway and a potential target for NSCLC therapy.
... Many cellular processes are tightly controlled by the post-translational modifications (PTMs) of target proteins, and as one type of PTM, protein ubiquitination is involved in many physiological events including DNA repair, cell-cycle control, stress response, immune response, endocytosis, and vesicle trafficking (Clague et al., 2012;Isono and Nagel, 2014;Pickart, 2004;Vierstra, 2009;Zhou et al., 2017). Generally, ubiquitination, the covalent attachment of the small protein modifier ubiquitin to a substrate protein (Chou et al., 2017), and deubiquitination, the removal of ubiquitin, often antagonistically regulate the ubiquitination level of target proteins, and are strictly controlled at multiple steps during cellular processes (Katz et al., 2010;Neutzner and Neutzner, 2012;Vierstra, 2009;Wilkinson, 2000). ...
... Except for the JAMM, which are zinc-dependent metalloproteases, all DUB belong to cysteine proteases (Komander et al., 2009;Nijman et al., 2005;Reyes-Turcu et al., 2009). Generally, DUBs recognize substrates anchored with monoubiquitin and polyubiquitin chain assemblies, and selectively cleave them (Katz et al., 2010;Neutzner and Neutzner, 2012;Sahtoe and Sixma, 2015;Soboleva and Baker, 2004;Wilkinson, 2000;Wolberger, 2014). Factually, protein ubiquitination which is essential for many cellular processes is dynamically regulated both by UBEs and DUBs (Fig. 1). ...
Article
Full-text available
As one type of deubiquitinases (DUBs), ubiquitin-specific proteases (UBPs) play an extensive and significant role in plant life involving the regulation of plant development and stress responses. However, comprehensive studies are still needed to determine the functional mechanisms, which are largely unclear. Here, we summarized recent progress of plant UBPs' functional partners, particularly the molecular mechanisms by which UBPs work with their partners. We believe that functional analyses of UBPs and their partners will provide new insights into protein deubiquitination and lead to a better understanding of the physiological roles of UBPs in plants.
... The lysine residue of one ubiquitin and the c-terminus of the following ubiquitin is where the hydrolysis occurs. DUBs perform a variety of tasks in the UPS, such as processing ubiquitin recycling, precursors, controlling non-degradation ubiquitin signals, and controlling protein stability [55,56]. Ubiquitin precursors are cleaved by DUBs consisting multiple ubiquitin as tandem repeats or ubiquitin are fused with the L40 and S27 ribosomal protein at the N-terminus [57]. ...
Article
The ubiquitin proteasomal system (UPS) represents a highly regulated protein degradation pathway essential for maintaining cellular homeostasis. This system plays a critical role in several cellular processes, which include DNA damage repair, cell cycle checkpoint control, and immune response regulation. Recently, the UPS has emerged as a promising target for cancer therapeutics due to its involvement in oncogenesis and tumor progression. Here we aim to summarize the key aspects of the UPS and its significance in cancer therapeutics. We begin by elucidating the fundamental components of the UPS, highlighting the role of ubiquitin, E1-E3 ligases, and the proteasome in protein degradation. Furthermore, we discuss the intricate process of ubiquitination and proteasomal degradation, emphasizing the specificity and selectivity achieved through various signaling pathways. The dysregulation of the UPS has been implicated in cancer development and progression. Aberrant ubiquitin-mediated degradation of key regulatory proteins, such as tumor suppressors and oncoproteins, can lead to uncontrolled cell proliferation, evasion of apoptosis, and metastasis. We outline the pivotal role of the UPS in modulating crucial oncogenic pathways, including the regulation of cyclins, transcription factors, Replication stress components and DNA damage response. The increasing recognition of the UPS as a target for cancer therapeutics has spurred the development of small molecules, peptides, and proteasome inhibitors with the potential to restore cellular balance and disrupt tumor growth. We provide an overview of current therapeutic strategies aimed at exploiting the UPS, including the use of proteasome inhibitors, deubiquitinating enzyme inhibitors, and novel E3 ligase modulators. We further discuss novel emerging strategies for the development of next-generation drugs that target proteasome inhibitors. Exploiting the UPS for cancer therapeutics offers promising avenues for developing innovative and effective treatment strategies, providing hope for improved patient outcomes in the fight against cancer.
... Among them, protein ubiquitination is an important post-translational modification (Komander, 2009). Ubiquitination is the covalent binding of ubiquitin to substrate proteins, while deubiquitination is the detachment of ubiquitin from substrate proteins, which usually antagonistically modulates the level of ubiquitination of target proteins and is tightly controlled (Katz et al., 2010;Neutzner and Neutzner, 2012;Vierstra, 2009;Wilkinson, 1997). The ubiquitination process requires the joint participation of E1, E2 and E3 proteins (Schulman and Harper, 2009;Vierstra, 2009;Ye and Rape, 2009). ...
Article
Low-temperature chilling injury poses a formidable threat to plant growth, development, and crop productivity. MYB transcription factors have emerged as key players, orchestrating a symphony of positive regulation in the realms of cold tolerance and flavonoid accumulation. However, the intricate regulatory mechanisms underlying safflower's response to such stressors still remain unclear. In the present study, a putative CtMYB63 was discovered to positively regulate low-temperature tolerance and hydroxysafflor yellow A accumulation in saf-flower. CtMYB63 exhibited up-regulation in the face of low-temperature stress. Similarly, the silencing of CtMYB63 via RNAi showed down-regulation of key genes involved in cold stress pathway emphasized their susceptibility to low temperature stress unlike overexpressed and wild-type plants. In addition, the interaction between CtMYB63 and a deubiquitin enzyme (CtUCH1) was also confirmed through yeast two-hybrid analysis. Remarkably, the overexpression of CtUCH1 in safflower rebounded the positive effects on low-temperature tolerance, further corroborating its regulatory prowess. Interestingly, while the accumulation of Hydroxysaf-flor yellow A remained unaffected in CtUCH1 overexpressed plants, it showed positive impact on cold tolerance. Furthermore, the overexpression of CtUCH1 in MYB63 mutant Arabidopsis leads to the restoration of MYB63 function and enhance its ability to regulate downstream target genes. This restoration of MYB function highlighted the intricate interplay between CtUCH1 and CtMYB63 in shaping safflower's transcriptional landscape. The detrimental effects on cold tolerance and Hydroxysafflor yellow A accumulation became evident as the U-box family of E3 ubiquitin ligases, particularly CtU-box1, targeted CtMYB63 for ubiquitination. In this complex interplay, the deubiquitinating prowess of CtUCH1 emerged as a vital modulator, preserving the stability of CtMYB63 by counteracting the adverse effects of CtU-box1. By elucidating this novel regulatory module, consisting of CtMYB63-CtU-box1-CtUCH1, our study pioneers an innovative pathway for enhancing cold stress resilience and optimizing Hydroxysafflor yellow A accumulation in safflower. These findings not only deepen our understanding of safflower's intricate adaptive mechanisms in response to cold stress but also pave the way for the development of novel strategies in crop improvement and pharmaceutical applications.
... On the other hand, deubiquitination is the cleavage of Ub from substrate proteins with the help of deubiquitinating enzymes (DUBs), protecting the target protein from being degraded. However, in some cases, DUBs can facilitate substrate protein degradation [36,37]. Deubiquitination is associated with various cellular processes, such as the regulation of cell cycle [38], gene expression [39], the degradation of lysosome-and proteosome-dependant proteins [40,41], the pathogenesis of microbes [42,43], kinase activation [44,45], DNA repair machinery [46], etc. DUBs are classified into seven subgroups: ubiquitin-specific proteases (USPs), JAB1/MPN/Mov34 metalloenzyme, ovarian tumor proteases (OTUs), Josephin and JAB1/MPN+(MJP), MIU-containing novel DUB (MINDY), zinc finger-containing ubiquitin peptidase 1 (ZUP1), and ubiquitin C-terminal hydrolases (UCHs) [47][48][49]. ...
Article
As cancer continues to be one of the leading causes of death, various cancer treatments are being developed, from traditional surgery to the more recent emergence of target therapy. However, therapy resistance is a restricting problem that needs to be overcome. Henceforth, the field of research shifts to new plausible drug targets, among which is the ubiquitin-proteasome system. This review is focused on the ubiquitin carboxyl-terminal hydrolase (UCH) protease family, which are members of Deubiquitinating enzymes (DUBs), specifically Ubiquitin carboxyl-terminal hydrolase L3 (UCHL3). DUBs regulate a broad array of regulatory processes, including cell-cycle progression, tissue development, and differentiation. DUBs are classified into seven subfamilies, including ubiquitin-specific proteases (USPs), JAB1/MPN/Mov34 metalloenzyme, ovarian tumor proteases (OTUs), Josephin and JAB1/MPN+(MJP), MIU-containing novel DUB (MINDY), zinc finger-containing ubiquitin peptidase 1 (ZUP1), and ubiquitin C-terminal hydrolases (UCHs). Having a significant role in tumorigenesis, UCHL3 is thus emerging as a therapeutic target. Knowing its involvement in cancer, it’s important to understand the structure of UCHL3, its substrate specificity, and its interaction to pave the way for the development of potential inhibitors. This review covers several directions of proteasome inhibitors drug discovery and small molecule inhibitors development.
... Nevertheless, in some cases, DUBs can also promote substrate degradation. 4,5 The balance between ubiquitination and deubiquitination is indispensable for all kinds of biological processes. 6,7 The DUB enzymes identified are divided into five subfamilies, [8][9][10][11] one of which is the ubiquitin C-terminal hydrolase (UCH) family. ...
Article
Full-text available
Cancer stem cells (CSCs) exhibit highly aggressive and metastatic features and resistance to chemotherapy and radiotherapy. Aryl hydrocarbon receptor (AhR) expression varies among non-small cell lung cancers (NSCLCs), and the mechanisms that support abnormal AhR expression in CSCs remain elusive. Here, we identified ubiquitin carboxyl terminal hydrolase L3 (UCHL3), a DUB enzyme in the UCH protease family, as a bona fide deubiquitylase of the AhR in NSCLC. UCHL3 was shown to interact with, deubiquitylate, and stabilize AhR in a manner dependent on its deubiquitylation activity. Moreover, we showed that UCHL3 promotes the stem-like characteristics and potent tumorigenic capacity of NSCLC cells. UCHL3 increased AhR stability and the binding of AhR to the promoter regions of the “stemness” genes ATP-binding cassette subfamily G member 2 (ABCG2), KLF4, and c-Myc. Depletion of UCHL3 markedly downregulated the “stemness” genes ABCG2, KLF4, and c-Myc, leading to the loss of self-renewal and tumorigenesis in NSCLCs. Furthermore, the UCHL3 inhibitor TCID induced AhR degradation and exhibited significantly attenuated efficacy in NSCLC cells with stem cell-like properties. Additionally, UCHL3 was shown to indicate poor prognosis in patients with lung adenocarcinoma. In general, our results reveal that the UCHL3 deubiquitylase is pivotal for AhR protein stability and a potential target for NSCLC-targeted therapy.
... Meanwhile, ubiquitination can be reversed by an opposite process, called deubiquitination, which removes Ub from target proteins and cooperates with Ub to regulate the ubiquitination levels of target proteins [4,8]. To date, deubiquitination has been found to be governed by deubiquitinases/deubiquitinating enzymes (DUBs), which generally affect the activities, stabilities, and fates of the target proteins [9,10]. ...
Article
Full-text available
The largest group of deubiquitinases—ubiquitin-specific proteases (UBPs)—perform extensive and significant roles in plants, including the regulation of development and stress responses. A comprehensive analysis of UBP genes has been performed in Arabidopsis thaliana, but no systematic study has been conducted in moso bamboo (Phyllostachys edulis). In this study, the genome-wide identification, classification, gene, protein, promoter region characterization, divergence time, and expression pattern analyses of the UBPs in moso bamboo were conducted. In total, 48 putative UBP genes were identified in moso bamboo, which were divided into 14 distinct subfamilies in accordance with a comparative phylogenetic analysis using 132 full-length protein sequences, including 48, 27, 25, and 32 sequences from moso bamboo, A. thaliana, rice (Oryza sativa), and purple false brome (Brachypodium distachyon), respectively. Analyses of the evolutionary patterns and divergence levels revealed that the PeUBP genes experienced a duplication event approximately 15 million years ago and that the divergence between PeUBP and OsUBP occurred approximately 27 million years ago. Additionally, several PeUBP members were significantly upregulated under abscisic acid, methyl jasmonate, and salicylic acid treatments, indicating their potential roles in abiotic stress responses in plants.
... In some cases, deubiquitinating enzymes (DUBs), which are capable of removing Ub from protein substrates, protect proteins from degradation and release free Ub for recycling. However, in other cases, DUBs also enhance substrate degradation [9][10][11]. Protein ubiquitination and deubiquitination participate in a number of biological processes, including cell growth and differentiation, transcriptional regulation, and oncogenesis. ...
Article
Full-text available
Protein ubiquitination and deubiquitination participate in a number of biological processes, including cell growth, differentiation, transcriptional regulation, and oncogenesis. Ubiquitin C-terminal hydrolases (UCHs), a subfamily of deubiquitinating enzymes (DUBs), includes four members: UCH-L1/PGP9.5 (protein gene product 9.5), UCH-L3, UCHL5/UCH37, and BRCA1-associated protein-1 (BAP1). Recently, more attention has been paid to the relationship between the UCH family and malignancies, which play different roles in the progression of different tumors. It remains controversial whether UCHL1 is a tumor promoter or suppressor. UCHL3 and UCH37 are considered to be tumor promoters, while BAP1 is considered to be a tumor suppressor. Studies have showed that UCH enzymes influence several signaling pathways that play crucial roles in oncogenesis, tumor invasion, and migration. In addition, UCH families are associated with tumor cell sensitivity to therapeutic modalities. Here, we reviewed the roles of UCH enzymes in the development of tumors, highlighting the potential consideration of UCH enzymes as new interesting targets for the development of anticancer drugs.
Article
Full-text available
Ubiquitination is essential for plant growth and development. Deubiquitination cooperates with ubiquitination to regulate the ubiquitination levels of target proteins. The ubiquitin-specific protease (UBP) family is the largest group of deubiquitinases (DUBs), which perform extensive and significant roles in eukaryotic organisms. However, the UBP genes in wheat ( TaUBPs ) are not identified, and the functions of TaUBPs are unknown. The present study identified 97 UBP genes in the whole genome of T . aestivum . These genes were divided into 15 groups and non-randomly distributed on chromosomes of T . aestivum . Analyses of evolutionary patterns revealed that TaUBPs mainly underwent purification selection. The studies of cis -acting regulatory elements indicated that they might be involved in response to hormones. Quantitative real-time PCR (qRT-PCR) results showed that TaUBPs were differentially expressed in different tissues. Besides, several TaUBPs were significantly up-regulated when plants were treated with salicylic acid (SA), implying that these DUBs may play a role in abiotic stress responses in plants and few TaUBPs displayed differential expression after viral infection. Furthermore, TaUBP1A.1 ( TraesCS1A02G432600.1 ) silenced by virus-induced gene silencing (VIGS) facilitates Chinese wheat mosaic virus (CWMV) infection in wheat, indicating that TaUBP1A.1 may be involved in a defense mechanism against viruses. This study comprehensively analyzed the UBP gene family in wheat and provided a basis for further research of TaUBPs functions in wheat plant response to viral infection.
Thesis
Les protéasomes sont de grands assemblages macromoléculaires ubiquitaires composés d’un complexe catalytique 20S et d’une particule régulatrice comprenant un module AAA-ATPase. Cette machine cellulaire est chargée de dégrader sélectivement les protéines intracellulaires pour permettre le renouvellement du protéome, éliminer les protéines défectueuses et contrôler de nombreuses fonctions biologiques. Le travail de cette thèse avait pour objectif de mettre à jour les mécanismes qui permettent aux complexes AAA-ATPase de déplier sélectivement les protéines substrat et de les transférer à la particule 20S, dans laquelle elles sont détruites. Pour cela, une approche novatrice a été utilisée en combinant la diffusion de neutrons aux petits angles résolue en temps (TR-SANS) avec la spectroscopie de fluorescence permettant de suivre l’activité biochimique. Le protéasome de l’archée hyperthermophile Methanocaldococcus jannaschii, a été utilisé comme système modèle. Il est composé de la protéase 20S et de la particule régulatrice AAA-ATPase PAN. Un variant de la protéine fluorescente GFP a été utilisé comme protéine substrat.Les données obtenues montrent que l’activité de dépliement de PAN génère des formes de GFP dénaturée formant des agrégats. En revanche, l’association avec la particule 20S prévient la formation de ces espèces et indique qu’une fois le dépliement d’une protéine par PAN engagé, les processus de transfert dans le complexe 20S et de dégradation sont étroitement couplés. L’analyse des spectres de diffusion neutronique du substrat GFP montrent que la population de GFP native disparait rapidement au profit des peptides générés par la protéase 20S, comme confirmé par une analyse en spectrométrie de masse. Cela démontre le caractère hautement processif du protéasome. Enfin, deux modes d'action de PAN ont été mis en évidence selon la quantité de protéines à dégrader par rapport au protéasome PAN-20S. Ces travaux permettent de valider expérimentalement un des modèles de fonctionnement du protéasome préalablement proposés et soulignent l’importance d’un contrôle de l’association des protéasomes in vivo. Cette étude met également en valeur l’intérêt de la technique TR-SANS pour étudier la dynamique fonctionnelle de grandes machines cellulaires.
Article
Full-text available
NF-κB transcription factors mediate the effects of pro-inflammatory cytokines such as tumour necrosis factor-α and interleukin-1β1. Failure to downregulate NF-κB transcriptional activity results in chronic inflammation and cell death, as observed in A20-deficient mice2. A20 is a potent inhibitor of NF-κB signalling, but its mechanism of action is unknown2. Here we show that A20 downregulates NF-κB signalling through the cooperative activity of its two ubiquitin-editing domains. The amino-terminal domain of A20, which is a de-ubiquitinating (DUB) enzyme of the OTU (ovarian tumour) family3, removes lysine-63 (K63)-linked ubiquitin chains from receptor interacting protein (RIP), an essential mediator of the proximal TNF receptor 1 (TNFR1) signalling complex4, 5. The carboxy-terminal domain of A20, composed of seven C2/C2 zinc fingers6, then functions as a ubiquitin ligase by polyubiquitinating RIP with K48-linked ubiquitin chains, thereby targeting RIP for proteasomal degradation. Here we define a novel ubiquitin ligase domain and identify two sequential mechanisms by which A20 downregulates NF-κB signalling. We also provide an example of a protein containing separate ubiquitin ligase and DUB domains, both of which participate in mediating a distinct regulatory effect.
Article
Full-text available
The spindle checkpoint prevents chromosome mis-segregation by delaying sister chromatid separation until all chromosomes have achieved bipolar attachment to the mitotic spindle. Its operation is essential for accurate chromosome segregation, whereas its dysregulation can contribute to birth defects and tumorigenesis. The target of the spindle checkpoint is the anaphase-promoting complex (APC), a ubiquitin ligase that promotes sister chromatid separation and progression to anaphase. Using a short hairpin RNA screen targeting components of the ubiquitin-proteasome pathway in human cells, we identified the deubiquitinating enzyme USP44 (ubiquitin-specific protease 44) as a critical regulator of the spindle checkpoint. USP44 is not required for the initial recognition of unattached kinetochores and the subsequent recruitment of checkpoint components. Instead, it prevents the premature activation of the APC by stabilizing the APC-inhibitory Mad2–Cdc20 complex. USP44 deubiquitinates the APC coactivator Cdc20 both in vitro and in vivo, and thereby directly counteracts the APC-driven disassembly of Mad2–Cdc20 complexes (discussed in an accompanying paper). Our findings suggest that a dynamic balance of ubiquitination by the APC and deubiquitination by USP44 contributes to the generation of the switch-like transition controlling anaphase entry, analogous to the way that phosphorylation and dephosphorylation of Cdk1 by Wee1 and Cdc25 controls entry into mitosis.
Article
Full-text available
We have identified a novel protein, BAP1, which binds to the RING finger domain of the Breast/Ovarian Cancer Susceptibility Gene product, BRCA1. BAP1 is a nuclear-localized, ubiquitin carboxy-terminal hydrolase, suggesting that deubiquitinating enzymes may play a role in BRCA1 function. BAP1 binds to the wild-type BRCA1-RING finger, but not to germline mutants of the BRCA1-RING finger found in breast cancer kindreds. BAP1 and BRCA1 are temporally and spatially co-expressed during murine breast development and remodeling, and show overlapping patterns of subnuclear distribution. BAP1 resides on human chromosome 3p21.3; intragenic homozygous rearrangements and deletions of BAP1 have been found in lung carcinoma cell lines. BAP1 enhances BRCA1-mediated inhibition of breast cancer cell growth and is the first nuclear-localized ubiquitin carboxy-terminal hydrolase to be identified. BAP1 may be a new tumor suppressor gene which functions in the BRCA1 growth control pathway.
Article
Full-text available
PHLPP1 belongs to a novel family of Ser/Thr protein phosphatases that serve as tumor suppressors by negatively regulating Akt signaling. Our recent studies have demonstrated that loss of PHLPP expression occurs at high frequency in colorectal cancer. In this study, we identified PHLPP1 as a proteolytic target of a β-TrCP-containing Skp-Cullin 1-F-box protein (SCF) complex (SCFβ-TrCP) E3 ubiquitin ligase in a phosphorylation-dependent manner. Overexpression of wild-type but not ΔF-box mutant β-TrCP leads to decreased expression and increased ubiquitination of PHLPP1, whereas knockdown of endogenous β-TrCP has the opposite effect. In addition, we show that the β-TrCP-mediated degradation requires phosphorylation of PHLPP1 by casein kinase I and glycogen synthase kinase 3β (GSK-3β), and activation of the phosphatidylinositol 3-kinase/Akt pathway suppresses the degradation of PHLPP1 by inhibiting the GSK-3β activity. Furthermore, expression of a degradation-deficient PHLPP1 mutant in colon cancer cells results in a more effective dephosphorylation of Akt and inhibition of cell growth. Taken together, our findings demonstrate a key role for β-TrCP in controlling the level of PHLPP1, and activation of Akt negatively regulates this degradation process.
Article
Full-text available
The scattering response of epithelial cells to activation of the Met receptor tyrosine kinase represents one facet of an "invasive growth" program. It is a complex event that incorporates loss of cell-cell adhesion, morphological changes, and cell motility. Ubiquitination is a reversible posttranslational modification that may target proteins for degradation or coordinate signal transduction pathways. There are approximately 79 active deubiquitinating enzymes (DUBs) predicted in the human genome. Here, via a small interfering RNA (siRNA) library approach, we have identified 12 DUBs that are necessary for aspects of the hepatocyte growth factor (HGF)-dependent scattering response of A549 cells. Different phenotypes are evident that range from full loss of scattering, similar to receptor knockdown (e.g., USP30, USP33, USP47), to loss of cell-cell contacts even in the absence of HGF but defective motility (e.g., USP3, ATXN3L). The knockdowns do not incur defective receptor, phosphatidylinositol 3-kinase, or MAP kinase activation. Our data suggest widespread involvement of the ubiquitin system at multiple stages of the Met activation response, implying significant crosstalk with phosphorylation-based transduction pathways. Development of small-molecule inhibitors of particular DUBs may offer a therapeutic approach to contain metastasis.
Article
Full-text available
Obesity results from the dysregulation of energy balance throughout the entire body. Although the ubiquitin system participates in many cellular processes, its contribution to the balance of energy in the body remains poorly understood. Here, we show that ubiquitin C-terminal hydrolase (UCH)-L3, one of the deubiquitinating enzymes, contributes to the regulation of metabolism. Uchl3(-/-) mice displayed a reduction of adipose tissue mass and were protected against high-fat diet (HFD)-induced obesity and insulin resistance. Uchl3(-/-) mice given both a normal chow and an HFD had an increased whole-body energy expenditure accounting for the reduction of adipose tissue mass. Activation of AMP-activated protein kinase (AMPK) in skeletal muscle has been reported to increase fatty acid beta-oxidation, leading to the elevation of the whole-body energy expenditure. Consistently, increased activation of AMPK and fatty acid beta-oxidation was observed in skeletal muscle of Uchl3(-/-) mice. Mouse embryonic fibroblasts derived from Uchl3(-/-) mice also showed increased activation of AMPK, indicating that UCH-L3 is involved in a cell-autonomous down-regulation of AMPK. These results suggest a role for UCH-L3 in the regulation of AMPK activity and whole-body energy metabolism.
Article
Full-text available
Ubiquitylation is a reversible protein modification that is implicated in many cellular functions. Recently, much progress has been made in the characterization of a superfamily of isopeptidases that remove ubiquitin: the deubiquitinases (DUBs; also known as deubiquitylating or deubiquitinating enzymes). Far from being uniform in structure and function, these enzymes display a myriad of distinct mechanistic features. The small number (<100) of DUBs might at first suggest a low degree of selectivity; however, DUBs are subject to multiple layers of regulation that modulate both their activity and their specificity. Due to their wide-ranging involvement in key regulatory processes, these enzymes might provide new therapeutic targets.
Article
Deubiquitinating enzymes (Dubs) function to remove covalently attached ubiquitin from proteins, thereby controlling substrate activity and/or abundance. For most Dubs, their functions, targets, and regulation are poorly understood. To systematically investigate Dub function, we initiated a global proteomic analysis of Dubs and their associated protein complexes. This was accomplished through the development of a software platform called CompPASS, which uses unbiased metrics to assign confidence measurements to interactions from parallel nonreciprocal proteomic data sets. We identified 774 candidate interacting proteins associated with 75 Dubs. Using Gene Ontology, interactome topology classification, subcellular localization, and functional studies, we link Dubs to diverse processes, including protein turnover, transcription, RNA processing, DNA damage, and endoplasmic reticulum-associated degradation. This work provides the first glimpse into the Dub interaction landscape, places previously unstudied Dubs within putative biological pathways, and identifies previously unknown interactions and protein complexes involved in this increasingly important arm of the ubiquitin-proteasome pathway.
Article
The Wnt/beta-catenin signalling pathway has important roles in normal cellular proliferation, development and angiogenesis. Many malignant transformations, including sporadic colorectal tumours, are caused by constitutive activation of the Wnt route due to mutations in the tumour suppressor protein adenomatous polyposis coli (APC) or the beta-catenin oncogene, ultimately resulting in reduced beta-catenin degradation by the ubiquitin (Ub) proteasome system (UPS). The COP9 signalosome (CSN) regulates the UPS by controlling cullin-RING Ub ligases (CRLs). We show here that the CSN and the beta-catenin destruction complex cooperate in targeting beta-catenin for degradation by the UPS. Together with the CRL that ubiquitinates beta-catenin, they form a supercomplex responsible for beta-catenin degradation. Wnt3A, glycogen synthase kinase 3beta inhibitors or mutation of CSN-mediated deneddylation induce the disassembly of the supercomplex and the accumulation of beta-catenin. Likewise, downregulation of the CSN in HeLa cells leads to retarded degradation of beta-catenin. Additionally, we found that the knockdown of the CSN causes accelerated proteolysis of APC, an essential component of the beta-catenin destruction complex, which is degraded by the UPS as beta-catenin. We show here that APC is stabilised by the Ub-specific protease 15 (USP15) associated with the CSN. This is demonstrated by over-expression of siRNA oligonucleotides against USP15 or by over-expression of an USP15 mutant, which is unable to degrade poly-Ub chains. Thus, the CSN controls the Wnt/beta-catenin signalling by assisting the assembly of beta-catenin-degrading supercomplexes by deneddylation and, simultaneously, by stabilising APC via CSN-associated USP15. The CSN regulates the balance between beta-catenin and APC. Disturbance of this balance can cause cancer by driving cell transformation, tumour angiogenesis and metastasis. A model is provided that proposes a role of CSN-mediated deneddylation in the formation of the beta-catenin-degrading supercomplex and the protection of complex-bound APC via CSN-associated USP15.
Article
The proteasome is an intricate molecular machine, which serves to degrade proteins following their conjugation to ubiquitin. Substrates dock onto the proteasome at its 19-subunit regulatory particle via a diverse set of ubiquitin receptors and are then translocated into an internal chamber within the 28-subunit proteolytic core particle (CP), where they are hydrolyzed. Substrate is threaded into the CP through a narrow gated channel, and thus translocation requires unfolding of the substrate. Six distinct ATPases in the regulatory particle appear to form a ring complex and to drive unfolding as well as translocation. ATP-dependent, degradation-coupled deubiquitination of the substrate is required both for efficient substrate degradation and for preventing the degradation of the ubiquitin tag. However, the proteasome also contains deubiquitinating enzymes (DUBs) that can remove ubiquitin before substrate degradation initiates, thus allowing some substrates to dissociate from the proteasome and escape degradation. Here we examine the key elements of this molecular machine and how they cooperate in the processing of proteolytic substrates.