ArticlePDF AvailableLiterature Review

Signalling pathways and the regulation of SUMO conjugation

Authors:

Abstract and Figures

The modification of proteins by SUMO (small ubiquitin-related modifier) conjugation is becoming increasingly recognized as an important regulatory event. Protein SUMOylation can control a whole range of activities, including subcellular localization, protein-protein interactions and enzymatic activity. However, the SUMOylation process can itself be controlled. In the present review, the mechanisms through which protein SUMOylation is regulated are discussed, with particular emphasis on the impact of signalling pathways. A major point of regulation of the SUMO pathway is through targeting the E3 ligases, and a number of different ways to achieve this have been identified. More generally, the MAPK (mitogen-activated protein kinase) pathways represent one way through which SUMOylation of specific proteins is controlled, by using molecular mechanisms that at least in part also function by modifying the activity of SUMO E3 ligases. Further intricacies in signalling pathway interactions are hinted at through the growing number of examples of cross-talk between different post-translational modifications and SUMO modification.
Content may be subject to copyright.
1414 Biochemical Society Transactions (2007) Volume 35, part 6
Signalling pathways and the regulation of SUMO
modification
B. Guo*, S.-H. Yang*, J. Witty* and A.D. Sharrocks*
1
*Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, U.K.
Abstract
The modification of proteins by SUMO (small ubiquitin-related modifier) conjugation is becoming increasingly
recognized as an important regulatory event. Protein SUMOylation can control a whole range of activities,
including subcellular localization, protein–protein interactions and enzymatic activity. However, the
SUMOylation process can itself be controlled. In the present review, the mechanisms through which protein
SUMOylation is regulated are discussed, with particular emphasis on the impact of signalling pathways.
A major point of regulation of the SUMO pathway is through targeting the E3 ligases, and a number of
different ways to achieve this have been identified. More generally, the MAPK (mitogen-activated protein
kinase) pathways represent one way through which SUMOylation of specific proteins is controlled, by using
molecular mechanisms that at least in part also function by modifying the activity of SUMO E3 ligases.
Further intricacies in signalling pathway interactions are hinted at through the growing number of examples
of cross-talk between different post-translational modifications and SUMO modification.
Introduction
The SUMO (small ubiquitin-related modifier) modification
pathway shows many parallels with the ubiquitin pathway.
Indeed, many of the pathway components show structural
and functional homology (reviewed in [1,2]). This is
illustrated by the fact that SUMO itself is a small polypeptide
that is structurally related to ubiquitin. The SUMO
pathway contains three main enzymatic components,
the E1 activating enzyme [SAE (SUMO1-activating
enzyme) 1/SAE2 also named Aos1/Uba2 (ubiquitin-
activating enzyme 2)], the E2-conjugating enzyme Ubc9
(ubiquitin-conjugating enzyme 9) and a number of E3
ligases that can facilitate the SUMOylation process in a
substrate-specific manner (Figure 1). SUMOylation can
be reversed through the proteolytic action of members
of the SENP [sentrin (also known as SUMO)-specific
protease] family. SUMOylation takes place on lysine
residues that are usually embedded within the core
consensus motif KXE [3]. However, other more exten-
ded motifs have been proposed, including the synergy control
motif [4] (characterized by proline residues in the flanking
regions) and the NDSM (negatively charged amino acid-
Key words: E3 ligase, mitogen-activated protein kinase (MAPK) pathway, protein inhibitor of
activated STAT (PIAS), phosphorylation-dependent SUMOylation motif (PDSM), small ubiquitin-
related modifier (SUMO), SUMOylation.
Abbreviations used: AIB1, amplified in breast cancer 1; CDK, cyclin-dependent kinase;
ERK, extracellular-signal-regulated kinase; ETS, E twenty-six; Elk-1, ETS-like kinase 1; HIC-1,
hypermethylated in cancer 1; HIPK2, homeodomain-interacting protein kinase 2; IκB, inhibitor
of nuclear factor κ B; IKKα,IκBkinaseα; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated
protein kinase; MEK-1, MAPK/ERK kinase 1; MEF2, myocyte enhancer factor 2; NDSM, negatively
charged amino acid-dependent SUMOylation motif; NF-κB, nuclear factor κB; NEMO, NF-κB
essential modulator; PEA3, polyoma enhancer activator 3; SAE, SUMO1-activating enzyme; STAT,
signal transducer and activator of transcription; PIAS, protein inhibitor of activated STAT; SUMO,
small ubiquitin-related modifier; PDSM, phosphorylation-dependent SUMO modification; RanBP2,
Ran-binding protein 2; Ubc9, ubiquitin-conjugating enzyme 9.
1
To whom correspondence should be addressed (email a.d.sharrocks@manchester.ac.uk).
dependent SUMOylation motif) (characterized by clusters
of acidic residues in the downstream region) [5]. These
extended motifs provide further specificity and enhanced
intrinsic propensities for SUMOylation. Further variations
on these motifs can impart regulatory properties (see below).
However, while most of the substrates contain the core
KXE motif, a significant number do not, and in those cases,
other structural constraints are thought to be important.
The functions attributed to SUMO modification differ
significantly from those identified for ubiquitin, which
predominantly targets proteins for degradation. Indeed,
SUMO modification has been shown to exhibit a number
of disparate functions depending on the protein substrates
(reviewed in [1,2,6]). One of the major roles emerging for
SUMOylation is in controlling the output from transcrip-
tional regulatory proteins, and in particular in mediating
transcriptional repression (reviewed in [7]). However, roles
in recombination, DNA repair and a growing number of
cytoplasmic processes have been uncovered (reviewed in [ 6]).
Protein SUMOylation is not a static process. Indeed
SUMOylation appears very dynamic, with typically only a
small proportion of any protein being modified. The latter
observation has been suggested to equate to a role of SUMO
in establishing a particular state that is then maintained
through the action of other proteins and/or modifications
(reviewed in [1]). Dynamic SUMOylation is observed in
many situations, but is clearly demonstrated during circadian
clock control where cyclical SUMOylation of BMAL1
(brain and muscle Arnt-like protein-1) parallels its rhythmic
activation [8]. A number of ways of regulating protein
SUMOylation have been discovered (reviewed in [9]). Here,
we describe some of the latest advances in this area, with a
particular focus on the role of signalling pathways in the regu-
lation of E3 ligases and the role of MAPK (mitogen-activated
C
The Authors Journal compilation
C
2007 Biochemical Society
Biochemical Society Transactions www.biochemsoctrans.org
Regulation of Protein Function by SUMO Modification 1415
Figure 1 The SUMO pathway and points of regulation
Components of the SUMO pathway are shown as ovals, and a putative
substrate as a rectangle. Only one E1 (SAE1/SAE2) and one E2 (Ubc9)
enzyme exist but there are multiple potential E3 ligases. Three possible
points of regulation of the pathway are shown: through inactivating the
E1 SUMO activating enzyme (I), inactivating the E2 SUMO conjugating
enzyme (II), or changing the activity of the E3 ligases (III) (see the text
for details).
protein kinase) pathways in controlling protein SUMOy-
lation status.
Global mechanisms for SUMOylation
control
Protein SUMOylation can be regulated in a substrate-specific
manner (see below) or in a more global way. Typically,
global SUMOylation is controlled by targeting one of the
core SUMO pathway components. However, control exerted
over the whole pathway has also been observed. For example,
studies in keratinocytes demonstrated that the whole SUMO
pathway is transiently up-regulated during differentiation,
thereby causing changes in the global patterns of protein
SUMOylation [10]. It is also well documented that various
stress stimuli cause changes in global SUMOylation patterns
(reviewed in [9]). For example, various cellular stresses,
including heat shock, cause global increases in SUMOylation
in mammalian cells [11]. Conversely, reductions in global
SUMOylation levels can be seen and one way this is achieved
is through targeting the E2 enzyme Ubc9. Low doses of H
2
O
2
cause the reversible oxidation of the E2-conjugating enzyme
Ubc9 [12]. This oxidation event inactivates Ubc9 and causes
a global decrease in SUMOylation levels. Similar reductions
in global SUMOylation levels are seen upon infection with
the CELO (chicken embryo lethal orphan) adenovirus, and
this effect can be attributed to the Gam1 protein, which tar-
gets the E1 activating enzyme SAE1/SAE2 and the E2 Ubc9
for destruction [13]. In the case of viral infection, the loss of
SUMOylation is thought to benefit the virus due to the result-
ing de-repression of many transcriptional processes, thereby
providing a permissive environment for viral propagation.
MAPK pathways and regulation of
SUMOylation
One way in which SUMOylation can be controlled is
through the action of protein kinase cascades in response to
extracellular signals. In particular, such cascades potentially
provide specificity to controlling the SUMOylation of a
limited number of substrates under defined conditions.
The MAPK pathways are a common route through
which extracellular signals are transduced into intracellular
responses (reviewed in [14]). Different pathways can
transduce stress [JNK (c-Jun N-terminal kinase) and p38
pathways] or mitogenic [ERK (extracellular-signal-regulated
kinase) pathway] signals. A direct link between these
pathways and SUMOylation was first demonstrated by
the observation that activation of the ERK pathway caused
de-SUMOylation of the transcription factor Elk-1 [ETS
(E twenty-six)-like kinase 1] [15]. As SUMOylation levels
decrease, Elk-1 phosphorylation increases and permits Elk-1
to go from a transcriptionally repressive to a transcriptionally
active form. However, the link between MAPK activation
and Elk-1 de-SUMOylation remains obscure. More recently,
the E3 ligase PIAS [protein inhibitor of activated STAT
(signal transducer and activator of transcription)] xα has
been implicated in controlling this switch but appears to do
so independently of its SUMO ligase activity [16].
A growing number of links are now being made between
the MAPK pathways and protein SUMOylation. For
example, treatment of cells with 17β-oestradiol causes
up-regulation of the ERK MAPK pathway and enhanced
phosphorylation of the co-activator protein AIB1 (amplified
in breast cancer 1) {also known as RAC3 (receptor-associated
co-activator 3), ACTR (activator of thyroid and retinoic
acid receptors), SRC-3 (steroid receptor co-activator 3)
and p/CIP [p300/CBP (cAMP-response-element-binding
protein-binding protein)]}. This phosphorylation inversely
correlates with AIB1 SUMOylation, and mutational analysis
suggests mutual cross-regulation of phosphorylation and
SUMOylation [17]. It is however unclear how this occurs at
the molecular level as the SUMOylation sites are not close
to the phosphorylation sites in the primary sequence.
In contrast with the role of MAPK pathways in promoting
de-SUMOylation, it is also feasible that they might also
promote SUMOylation under some circumstances. Indeed,
recently, we have studied the role of ERK MAPK signalling
in potentially controlling SUMOylation of the ETS-
domain transcription factor PEA3 (polyoma enhancer
activator 3) and found that ERK signalling promotes PEA3
SUMOylation (B. Guo and A.D. Sharrocks, unpublished
work). Furthermore, SUMOylation appears to promote
transcriptional activation by PEA3; thus ERK signalling can
enhance the transactivation capacity of different ETS-domain
transcription factors through modifying their SUMOylation
status in an opposite manner (Figure 2). In the case of
Elk-1, loss of repressive SUMO enhances transactivation,
but for PEA3, increases in activating SUMO modifications
cause the same molecular effect, thereby providing two
C
The Authors Journal compilation
C
2007 Biochemical Society
1416 Biochemical Society Transactions (2007) Volume 35, part 6
Figure 2 MAPK-mediated control of SUMOylation
SUMOylation of transcription factors (TF) can enhance their repressive
activities (left) or increase their transactivation capacity (right). MAPKs
can potentially enhance the transactivation properties of transcription
factors through two diametrically opposed mechanisms: reduction of
SUMOylation levels (as observed for Elk-1 [15]) or increased SUMOylation
levels (as observed for PEA3; B. Guo and A.D. Sharrocks, unpublished
work).
different modes of regulatory logic to reach the same
endpoint.
Intriguingly, one of the first suggestions of a link between
MAPK signalling and the regulation of SUMOylation was
the observation that p38 signalling led to the increased
Smad4 SUMOylation [18]. T his increase in SUMOylation
was thought to be due to the up-regulation of the SUMO
E3 ligase PIASxβ at the transcriptional and protein levels.
In addition to increasing the levels of PIASx proteins, p38
signalling has also been directly implicated in modifying the
activity of these proteins [19]. These additional links between
the p38 pathway and E3 ligases were made during a study
of the effect of stress signalling on Elk-1 SUMOylation
levels. Whereas mitogenic signalling via the ERK MAPK
pathway causes de-SUMOylation, stress signalling via the
p38 pathway does not cause de-SUMOylation although
Elk-1 still becomes phosphorylated [19]. Interestingly, the
E3 ligase PIASxα plays a key role in both cases. Whereas
PIASxα promotes de-SUMOylation in response to ERK
pathway signalling, PIASxα is required to maintain Elk-1
SUMOylation upon p38 pathway activation. The switch
between these two modes of operation is through p38-
mediated phosphorylation of PIASxα. Thus PIASxα acts
as a molecular rheostat that controls the transcriptional
activation output from Elk-1 by either facilitating loss or
promoting retention of the repressive SUMO modification,
depending on the pathway that is activated.
Thus a common theme seems to be the involvement of
E3 ligases in controlling SUMOylation in the response to
signalling pathway activation.
E3 ligases as a focal point for regulation?
Unlike the ubiquitin pathway where multiple E2 proteins
exist, there is only a single E2 ligase in the SUMO pathway.
Thus any regulation of Ubc9 is likely to produce a global
response, as illustrated in the case of regulating Ubc9 activity
by H
2
O
2
[12]. Thus the E3 ligases represent an attractive
point for potential regulation. It is currently unclear
how many E3 ligases exist, but to date several have been
identified including PIASxα, PIASxβ, PIAS1, PIAS3, PIASy,
RanBP2 (Ran-binding protein 2), Pc2 and HDAC4 (histone
deacetylase 4) (reviewed in [1,20]). Several recent studies
have implicated these proteins as points for regulation.
NEMO (NF-κB essential modulator) is a regulator of
NF-κB (nuclear factor κB) activity, and NEMO itself is
SUMOylated in response to genotoxic agents, which in turn
leads to NF-κB activation. Recently, PIASy was identified
as the SUMO E3 ligase for NEMO, and genotoxic stress
promotes the interaction of NEMO and PIASy [21]. Thus an
important regulated interaction between a substrate and
an E3 ligase h as been defined, although it is currently unclear
how genotoxic stresses promote this interaction.
Another E3 ligase Pc2 has also been shown to be regulated
in response to DNA-damaging agents [22]. In this case,
direct molecular links have been established whereby HIPK2
(homeodomain-interacting protein kinase 2) is activated
following DNA damage, and this then phosphorylates
Pc2. The E3 ligase activity of phosphorylated Pc2 is
enhanced and one target is HIPK2 itself, and SUMOylation
promotes the ability of HIPK2 to act as a transcriptional
repressor. This provides a nice example of a feedback loop
where mutual regulatory modifications (SUMOylation and
phosphorylation) are exchanged between Pc2 and HIPK2.
Further interactions between signalling pathways and E3
ligases have been made with PIAS1, which is a direct target of
IKKα [IκB (inhibitor of NF-κB) kinase α] following expos-
ure of cells to pro-inflammatory stimuli [23]. Phosphoryl-
ation of PIAS1 promotes its ability to associate with pro-
moters and reduce the binding of other transcription factors
including STAT1 and NF-κB. Complex interactions between
SUMOylation and phosphorylation are suggested by the
observation that the E3 ligase activity of PIAS1 is needed
for phosphorylation by IKKα. However, the intrinsic ligase
activity of PIAS1 is not affected by phosphorylation. This
has several parallels with the Elk-1–PIASx interaction where
the SUMO E3 ligase is a target of a stress-activated signalling
pathway and the functional output is in promoting or inhib-
iting other binding events at the promoter rather than being
dependent on global changes in the E3 ligase activity [19].
A third class of E3 ligase, RanBP2, is also subject to
regulatory phosphorylation. In this case, CDKs (cyclin-
dependent kinases) act upon RanBP2 during the cell cycle,
but the functional consequences of this phosphorylation
event are currently unknown [24]. Furthermore, signalling
through the PKB (protein kinase B; also called Akt) pathway
leads to up-regulation of RanBP2 levels [25], suggesting that
RanBP2 is also subject to regulation at multiple levels.
Cross-talk between SUMO and other
post-translational modifications
SUMOylation takes place on lysine residues, which can
also be modified by other post-translational modifications,
including acetylation, methylation and ubiquitination.
This provides ample opportunity for regulatory cross-talk
between different pathways that culminate in different
modification events. Indeed several early studies on the
C
The Authors Journal compilation
C
2007 Biochemical Society
Regulation of Protein Function by SUMO Modification 1417
SUMO pathway highlighted the direct antagonism that exists
for SUMOylation and ubiquitination of the same residues
in IκBα [26]. Similarly, in PCNA (proliferating-cell nuclear
antigen), dynamic switching between SUMOylation and
ubiquitination of the same residues has been observed [27].
More recently, acetylation has been shown to antagonize
SUMOylation of the s ame lysine residues in substrates such
as MEF2 (myocyte enhancer factor 2) [20,28] and HIC-1
(hypermethylated in cancer 1) [29]. Moreover, in the case
of MEF2, phosphorylation appears to be a direct trigger
that permits switching between acetylated and SUMOylated
states [28]. Phosphorylation of MEF2 takes place on a serine
residue located three amino acids downstream from the
core SUMOylation motif, and phosphorylation promotes
SUMOylation [28,30,31]. The kinase thought to be involved
in this switch is CDK5, which recognizes the core SP motif
[30], and phosphorylation is reversed by the phosphatase
calcineurin [28]. A similar phosphorylation-dependent
switch controlling SUMOylation levels was also identified in
HSF-1 (heat-shock factor 1) [32] and led to the identification
of a panel of potential SUMO substrates that contain
this extended motif, KXEXXSP, and was named the
PDSM (phosphorylation-dependent SUMOylation motif)
(reviewed in [33]). Importantly, this motif is related to the
NDSM, as it appears that it is the negative charge imparted
by the phosphorylation event that is a key determinant
in enhancing substrate SUMOylation. Thus two types of
substrates can be identified in which SUMOylation is
constitutively enhanced due to the presence of an NDSM, or
inducibly controlled through the PDSM. A further variation
on this theme was suggested from work on HIC-1, where
the proline residues in the core motif, KXEP, were shown
to be important in controlling the acetylation–SUMOylation
switch on the lysine residue [29]. Thus different extended
SUMO motifs impart different regulatory potential to
the SUMOylation process.
The above examples therefore illustrate the potential
for regulatory cross-talk between different pathways and,
in the case of MEF2, direct links to cellular signalling
pathways have been made whereby these pathways control
substrate SUMOylation through controlling substrate
phosphorylation levels.
Perspectives
Our current view of SUMOylation is evolving from one
in which SUMO is a static modification or one that is
cycled as a default mechanism to one where, in many
cases, substrate SUMOylation is controlled by signalling
pathways. This control can be exerted directly through
phosphorylation-dependent switches on the substrate or
indirectly through control of the SUMO pathway itself. One
key area for regulation appears to be through the E3 ligases,
but equally regulation might occur at the level of the SUMO
proteases and this is likely to be an area for future studies.
The MAPK pathways have been linked to controlling
protein SUMOylation of several substrates, and it is tempting
to speculate that in some cases, novel direct regulatory links
will be made through the PDSM, KXEXXSP, as the SP
motif corresponds to the consensus site for this class of
kinase. Reciprocally, links between the SUMO pathway and
MAPK pathway activity can be envisaged as demonstrated
by the stimulatory effect that PIAS1 has on the JNK MAPK
pathway [34].
More recent discoveries such as the observation that
the protein tyrosine phosphatase PTP1B is regulated by
SUMOylation [35] suggest even more complex reciprocal
links between SUMOylation and signalling pathways.
Indeed, previous studies have demonstrated that the MAPK
pathway component MEK-1 (MAPK/ERK kinase 1) is
SUMOylated in Dictyostelium and t his SUMOylation
event is also regulated by MEK-1 phosphorylation by
upstream kinases [36]. In addition to classical signalling
pathways, nuclear hormone receptors can also influence
the output of signalling cascades by transrepression activity
at promoters. Ligand-dependent SUMOylation of nuclear
hormone receptors LXR (liver X receptor) and PPARγ
(peroxisome-proliferator-activated receptor γ ) enhances
their transrepressive properties and inhibits the expression of
genes that are usually responsive to inflammatory signalling
[37,38]. Thus, depending on the inflammatory signals
and nuclear hormone ligands present, then ultimately the
SUMOylation levels of nuclear hormone receptors control
the overall transcriptional response of the cell.
Future studies will undoubtedly uncover more intricacies
in the links between intracellular signalling pathways and
SUMOylation control. Potentially, these links may identify
important areas that could be targeted for therapeutic
intervention in diseases such as cancer where signalling
pathway wiring is often disrupted.
We apologize to our colleagues whose primary work was not
cited due to space constraints. The work in our laboratory on
protein SUMOylation is supported by the BBSRC (Biotechnology and
Biological Sciences Research Council), the Wellcome Trust and a
Royal Society-Wolfson award to A.D.S.
References
1 Hay, R.T. (2005) Mol. Cell 18,112
2 Johnson, E.S. (2004) Annu. Rev. Biochem. 73, 355–382
3 Rodriguez, M.S., Dargemont, C. and Hay, R.T. (2001) J. Biol. Chem. 276,
12654–12659
4 Iniguez-Lluhi, J.A. and Pearce, D. (2000) Mol. Cell. Biol. 20, 6040–6050
5 Yang, S.H., Galanis, A., Witty, J. and Sharrocks, A.D. (2006) EMBO J. 25,
5083–5093
6 Watts, F.Z. (2004) Semin. Cell Dev. Biol. 15, 211–220
7 Gill, G. (2005) Curr. Opin. Genet. Dev. 15, 536–541
8 Cardone, L., Hirayama, J., Giordano, F., Tamaru, T., Palvimo, J.J. and
Sassone-Corsi, P. (2005) Science 309, 1390–1394
9 Bossis, G. and Melchior, F. (2006) Cell Div. 1,13
10 Deyrieux, A.F., Rosas-Acosta, G., Ozbun, M.A. and Wilson, V.G. (2007)
J. Cell Sci. 120, 125–136
11 Saitoh, H. and Hinchey, J. (2000) J. Biol. Chem. 275, 6252–6258
12 Bossis, G. and Melchior, F. (2006) Mol. Cell 21, 349–357
13 Boggio, R., Colombo, R., Hay, R.T., Draetta, G.F. and Chiocca, S. (2004)
Mol. Cell 16, 549–561
C
The Authors Journal compilation
C
2007 Biochemical Society
1418 Biochemical Society Transactions (2007) Volume 35, part 6
14 Raman, M., Chen, W. and Cobb, M.H. (2007) Oncogene 26,
3100–3012
15 Yang, S.H., Jaffray, E., Hay, R.T. and Sharrocks, A.D. (2003) Mol. Cell 12,
63–74
16 Yang, S.H. and Sharrocks, A.D. (2005) EMBO J. 24, 2161–2171
17 Wu, H., Sun, L., Zhang, Y., Chen, Y., Shi, B., Li, R., Wang, Y., Liang, J.,
Fan, D., Wu, G. et al. (2006) J. Biol. Chem. 281, 21848–21856
18 Ohshima, T. and Shimotohno, K. (2003) J. Biol. Chem. 278,
50833–50842
19 Yang, S.H. and Sharrocks, A.D. (2006) Mol. Cell 22, 477–487
20 Zhao, X., Sternsdorf, T., Bolger, T.A., Evans, R.M. and Yao, T.P. (2005)
Mol. Cell. Biol. 25, 8456–8464
21 Mabb, A.M., Wuerzberger-Davis, S.M. and Miyamoto, S. (2006)
Nat. Cell Biol. 8, 986–993
22 Roscic, A., Moller, A., Calzado, M.A., Renner, F., Wimmer, V.C., Gresko, E.,
Ludi, K.S. and Schmitz, M.L. (2006) Mol. Cell 24, 77–89
23 Liu, B., Yang, Y., Chernishof, V., Loo, R.R., Jang, H., Tahk, S., Yang, R.,
Mink, S., Shultz, D., Bellone, C.J. et al. (2007) Cell 129, 903–914
24 Swaminathan, S., Kiendl, F., Korner, R., Lupetti, R., Hengst, L. and
Melchior, F. (2004) J. Cell Biol. 164, 965–971
25 Renner, O., Fominaya, J., Alonso, S., Blanco-Aparicio, C., Leal, J.F. and
Carnero, A. (2007) Carcinogenesis 28, 1418–1425
26 Desterro, J.M., Rodriguez, M.S. and Hay, R.T. (1998) Mol. Cell 2,
233–239
27 Hoege, C., Pfander, B., Moldovan, G.L, Pyrowolakis, G. and Jentsch, S.
(2002) Nature 419, 135–141
28 Shalizi, A., Gaudilliere, B., Yuan, Z., Stegmuller, J., Shirogane, T., Ge, Q.,
Tan, Y., Schulman, B., Harper, J.W. and Bonni, A. (2006) Science 311,
1012–1017
29 Stankovic-Valentin, N., Deltour, S., Seeler, J., Pinte, S., Vergoten, G.,
Guerardel, C., Dejean, A. and Leprince, D. (2007) Mol. Cell. Biol. 27,
2661–2675
30 Gregoire, S., Tremblay, A.M., Xiao, L., Yang, Q., Ma, K., Nie, J., Mao, Z.,
Wu, Z., Giguere, V. and Yang, X.J. (2006) J. Biol. Chem. 281, 4423–4433
31 Kang, J., Gocke, C.B. and Yu, H. (2006) BMC Biochem. 7,5
32 Hietakangas, V., Anckar, J., Blomster, H.A., Fujimoto, M., Palvimo, J.J.,
Nakai, A. and Sistonen, L. (2005) Proc. Natl. Acad. Sci. U.S.A. 103, 45–50
33 Yang, X.J. and Gregoire, S. (2006) Mol. Cell 23, 779–786
34 Liu, B. and Shuai, K. (2001) J. Biol. Chem. 276, 36624–36631
35 Dadke, S., Cotteret, S., Yip, S.C., Jaffer, Z.M., Haj, F., Ivanov, A.,
Rauscher, III, F., Shuai, K., Ng, T., Neel, B.G. and Chernoff, J. (2007)
Nat. Cell Biol. 9, 80–85
36 Sobko, A., Ma, H. and Firtel, R.A. (2002) Dev. Cell 2, 745–756
37 Pascual, G., Fong, A.L., Ogawa, S., Gamliel, A., Li, A.C., Perissi, V., Rose,
D.W., Willson, T.M., Rosenfeld, M.G. and Glass, C.K. (2005) Nature 437,
759–763
38 Ghisletti, S., Huang, W., Ogawa, S., Pascual, G., Lin, M.E., Willson, T.M.,
Rosenfeld, M.G. and Glass, C.K. (2007) Mol. Cell 25, 57–70
Received 25 July 2007
doi:10.1042/BST0351414
C
The Authors Journal compilation
C
2007 Biochemical Society
... One posttranslational mechanism that has emerged in recent years as an important regulatory mechanism of lipid metabolism is the conjugation of SUMO to target proteins. For example, the SUMOylation of key master regulators of lipid metabolism, the Sterol Regulatory Element Binding Proteins (SREBPs), coordinate lipid homeostasis with the physiological state of the organism (Figure 3) [1][2][3][4][5][6][7][8][9][10][11][12][13][14][15]. ...
... Within the many types of posttranscriptional modifications of proteins (PTMs), SUMO belongs to the superfamily of ubiquitinlike modifiers that have essential functions in eukaryotes. These functions include regulation of the cell cycle [3]; transcriptional and posttranscriptional control on mRNA levels [4]; DNA damage repair [5]; regulation of signal transduction [6]; and, indirectly, protein degradation [7]. The SUMO family members are short polypeptides of about 100 amino acids that are covalently conjugated to specific lysines in the sequence of their target substrates. ...
Article
Full-text available
One of the basic building blocks of all life forms are lipids—biomolecules that dissolve in nonpolar organic solvents but not in water. Lipids have numerous structural, metabolic, and regulative functions in health and disease; thus, complex networks of enzymes coordinate the different compositions and functions of lipids with the physiology of the organism. One type of control on the activity of those enzymes is the conjugation of the Small Ubiquitin-like Modifier (SUMO) that in recent years has been identified as a critical regulator of many biological processes. In this review, I summarize the current knowledge about the role of SUMO in the regulation of lipid metabolism. In particular, I discuss (i) the role of SUMO in lipid metabolism of fungi and invertebrates; (ii) the function of SUMO as a regulator of lipid metabolism in mammals with emphasis on the two most well-characterized cases of SUMO regulation of lipid homeostasis. These include the effect of SUMO on the activity of two groups of master regulators of lipid metabolism—the Sterol Regulatory Element Binding Protein (SERBP) proteins and the family of nuclear receptors—and (iii) the role of SUMO as a regulator of lipid metabolism in arteriosclerosis, nonalcoholic fatty liver, cholestasis, and other lipid-related human diseases.
... PTM by SUMOylation is a critical regulator of the molecular, cellular and systemic functions of proteins. Small ubiquitin-like modifier (SUMO) protein is a member of the ubiquitin-like (Ubl) family with a molecular weight of 10KDa and four isoforms (1, 2, 3, and 4) in humans [26][27][28][29]. SUMO1 and SUMO2 have 50% sequence identity, whereas SUMO2 and SUMO3 have 97% similarity. ...
... SUMO1 and SUMO2 have 50% sequence identity, whereas SUMO2 and SUMO3 have 97% similarity. Types 1, 2, and 3 are expressed ubiquitously, but SUMO4 is only found in the kidney, lymph nodes, and spleen [29][30][31][32][33]. ...
Article
Full-text available
Atherosclerosis is a progressive, inflammatory cardiovascular disorder characterized by the development of lipid-filled plaques within arteries. Endothelial cell dysfunction in the walls of blood vessels results in an increase in vascular permeability, alteration of the components of the extracellular matrix, and retention of LDL in the sub-endothelial space, thereby accelerating plaque formation. Epigenetic modification by SUMOylation can influence the surface interactions of target proteins and affect cellular functionality, thereby regulating multiple cellular processes. Small ubiquitin-like modifier (SUMO) can modulate NFκB and other proteins such as p53, KLF, and ERK5, which have critical roles in atherogenesis. Furthermore, SUMO regulates leukocyte recruitment and cytokine release and the expression of adherence molecules. In this review, we discuss the regulation by SUMO and SUMOylation modifications of proteins and pathways involved in atherosclerosis.
... edu. cn/) (Guo et al. 2007). The isoelectric point (pI) and molecular weight of the SUMO pathway proteins were analyzed using the ExPASy ProtParam tool (https:// web. ...
Article
Full-text available
Key message We have identified and analyzed 28 SUMO-pathway proteins from pigeonpea. Enhanced transcripts of pathway genes and increased SUMO conjugation under drought signifies the role of SUMO in regulating stress. Abstract Being a protein-rich and nutrient-dense legume crop, pigeonpea (Cajanus cajan) holds a vital position in a vegetarian meal. It is a resilient crop capable of striving in harsh climates and provides a means of subsistence to small-holding farmers. Nevertheless, extremes of water scarcity and drought conditions, especially during seedling and reproductive stages, remains a major issue severely impacting the growth and overall productivity of pigeonpea. Small ubiquitin-like modifier (SUMO), a post-translational modification system, plays a pivotal role in fortifying plants against stressful conditions by rapid reprogramming of molecular events. In this study, we have scanned the entire pigeonpea genome and identified 28 candidates corresponding to SUMO machinery components of pigeonpea. qRT-PCR analysis of different SUMO machinery genes validated their presence under natural conditions. The analysis of the promoters of identified SUMO machinery genes revealed the presence of abiotic stress-related cis-regulatory elements highlighting the potential involvement of the genes in abiotic stress responses. The transcript level analysis of selected SUMO machinery genes and global SUMO status of pigeonpea proteins in response to drought stress suggests an integral role of SUMO in regulating drought stress conditions in pigeonpea. Collectively, the work puts forward a detailed in silico analysis of pigeonpea SUMO machinery candidates and highlights the essential role of SUMOylation in drought stress responses. Being the first report on a pulse crop, the study will serve as a resource for devising strategies for counteracting drought stress in pigeonpea that can be further extended to other pulse crops.
... Small ubiquitin-like modifiers (SUMO), with a molecular weight of about 10 kDa, belong to the large family of ubiquitin-like (Ubl) proteins. There are four subtypes of SUMO in mammals, namely, SUMO1, SUMO2, SUMO3, and SUMO4 [29][30][31][32], of which SUMO2 and SUMO3 have about 97% similarity [33]. The subtypes SUMO1-SUMO3 are widely expressed, while SUMO4 is only expressed in the kidney, spleen, and lymph nodes [34][35][36]. ...
Article
Full-text available
Cardiovascular disease (CVD) is the leading cause of death in the world. The mechanism behind CVDs has been studied for decades; however, the pathogenesis is still controversial. Mitochondrial homeostasis plays an essential role in maintaining the normal function of the cardiovascular system. The alterations of any protein function in mitochondria may induce abnormal mitochondrial quality control and unexpected mitochondrial dysfunction, leading to CVDs. Posttranslational modifications (PTMs) affect protein function by reversibly changing their conformation. This review summarizes how common and novel PTMs influence the development of CVDs by regulating mitochondrial quality control. It provides not only ideas for future research on the mechanism of some types of CVDs but also ideas for CVD treatments with therapeutic potential.
... Many different mechanisms have been described for SUMOdependent repression in the literature (Gill, 2005;Hay, 2005;Guo et al, 2007;Lyst & Stancheva, 2007;Rosonina et al, 2017;Rosonina, 2019). We consider it most likely that SUMO regulates IRF2BP1 interactions in the context of specific promoters. ...
Article
Full-text available
Molecular switches are essential modules in signaling networks and transcriptional reprogramming. Here, we describe a role for small ubiquitin-related modifier SUMO as a molecular switch in epidermal growth factor receptor (EGFR) signaling. Using quantitative mass spectrometry, we compare the endogenous SUMO proteomes of HeLa cells before and after EGF stimulation. Thereby, we identify a small group of transcriptional coregulators including IRF2BP1, IRF2BP2, and IRF2BPL as novel players in EGFR signaling. Comparison of cells expressing wild type or SUMOylation-deficient IRF2BP1 indicates that transient deSUMOylation of IRF2BP proteins is important for appropriate expression of immediate early genes including dual specificity phosphatase 1 (DUSP1, MKP-1) and the transcription factor ATF3. We find that IRF2BP1 is a repressor, whose transient deSUMOylation on the DUSP1 promoter allows-and whose timely reSUMOylation restricts-DUSP1 transcription. Our work thus provides a paradigm how comparative SUMO proteome analyses serve to reveal novel regulators in signal transduction and transcription.
... Rights reserved. (Guo et al. 2007). Connections between SUMO and other SUMO lysine residues (K) contribute to the formation of substrate connections to the SUMO chain (poly-SUMOylation) (Pfander et al. 2005). ...
Article
Full-text available
Apoptosis and clearance of dead cells is highly evolutionarily conserved from nematode to humans, which is crucial to the growth and development of multicellular organism. Fail to remove apoptotic cells often lead to homeostasis imbalance, fatal autoimmune diseases, and neurodegenerative diseases. Small ubiquitin-related modifiers (SUMOs) modification is a post-translational modification of ubiquitin proteins mediated by the sentrin-specific proteases (SENPs) family. SUMO modification is widely involved in many cellular biological process, and abnormal SUMO modification is also closely related to many major human diseases. Recent researches have revealed that SUMO modification event occurs during apoptosis and clearance of apoptotic cells, and plays an important role in the regulation of apoptotic signaling pathways. This review summarizes some recent progress in the revelation of regulatory mechanisms of these pathways and provides some potential researching hotpots of the SUMO modification regulation to apoptosis.
Article
Full-text available
Acute myocardial infarction (AMI) is associated with high morbidity and mortality worldwide. Although early reperfusion is the most effective strategy to salvage ischemic myocardium, reperfusion injury can develop with the restoration of blood flow. Therefore, it is important to identify protection mechanisms and strategies for the heart after myocardial infarction. Recent studies have shown that multiple intracellular molecules and signaling pathways are involved in cardioprotection. Meanwhile, device-based cardioprotective modalities such as cardiac left ventricular unloading, hypothermia, coronary sinus intervention, supersaturated oxygen (SSO2), and remote ischemic conditioning (RIC) have become important areas of research. Herein, we review the molecular mechanisms of cardioprotection and cardioprotective modalities after ischemia–reperfusion injury (IRI) to identify potential approaches to reduce mortality and improve prognosis in patients with AMI.
Article
Background: Polyglutamine (polyQ) diseases are dominant neurodegenerative diseases caused by an expansion of the polyQ-encoding CAG repeats in the disease-causing gene. The length of the CAG repeats is the major determiner of the age at onset (AO) of polyQ diseases, including Huntington's disease (HD) and spinocerebellar ataxia type 3 (SCA3). Objective: We set out to identify common genetic variant(s) that may affect the AO of polyQ diseases. Methods: Three hundred thirty-seven patients with HD or SCA3 were enrolled for targeted sequencing of 583 genes implicated in proteinopathies. In total, 16 genes were identified as containing variants that are associated with late AO of polyQ diseases. For validation, we further investigate the variants of PIAS1 because PIAS1 is an E3 SUMO (small ubiquitin-like modifier) ligase for huntingtin (HTT), the protein linked to HD. Results: Biochemical analyses revealed that the ability of PIAS1S510G to interact with mutant huntingtin (mHTT) was less than that of PIAS1WT , resulting in lower SUMOylation of mHTT and lower accumulation of insoluble mHTT. Genetic knock-in of PIAS1S510G in a HD mouse model (R6/2) ameliorated several HD-like deficits (including shortened life spans, poor grip strength and motor coordination) and reduced neuronal accumulation of mHTT. Conclusions: Our findings suggest that PIAS1 is a genetic modifier of polyQ diseases. The naturally occurring variant, PIAS1S510G , is associated with late AO in polyQ disease patients and milder disease severity in HD mice. Our study highlights the possibility of targeting PIAS1 or pathways governing protein homeostasis as a disease-modifying approach for treating patients with HD. © 2021 International Parkinson and Movement Disorder Society.
Article
Background and aims: The physiological role of Rho-specific guanine nucleotide dissociation inhibitor (RhoGDI) in vascular remodeling remains unknown. We investigated the function of RhoGDI in angiotensin II (Ang II)-induced vascular remodeling in cultured human aortic vascular smooth muscle cells (HA-VSMCs) and in an Ang II-infusion vascular remodeling mouse model. Methods: For in vitro assays of HA-VSMCs, proliferation was assessed by BrdU and EdU assays and immunofluorescence analysis of ki-67 expression. RhoGDI1 and RhoGDI2 function and expression were assessed by RNAi, Western blotting and real-time RT-PCR. RhoGDI ubiquitination and SUMOylation levels were evaluated by co-immunoprecipitation and Western blotting. The functions of proteosomal-mediated degradation, ubiquitination, SUMOylation and Ang II receptors were assessed using specific inhibitors. To evaluate the in vivo effects of Ang II and RhoGDI, H & E staining, Masson's trichrome staining, and immunostaining were employed. Results: Ang II treatment of HA-VSMCs for 6 or 48 h promoted RhoGDI1 and RhoGDI2 protein degradation and reduced cell proliferation, which was reversed by proteosome inhibition. In contrast, treatment with Ang II for 12 or 24 h induced dose-dependent cell proliferation without affecting RhoGDI expression. RNA interference of either RhoGDI1 or RhoGDI2 blocked proliferation induced by 12 or 24 h treatment of Ang II. Moreover, Ang II-dependent degradation at 6 and 48 h correlated with RhoGDI ubiquitination and inversely correlated with RhoGDI SUMOylation and cell proliferation. Treatment with specific inhibitors suggests that ubiquitin and SUMO competitively bind to RhoGDI1 and RhoGDI2 to reciprocally regulate RhoGDI stability and HA-VSMC proliferation. Furthermore, inhibition of the Ang II receptor 1 (AT1 receptor), but not the Ang II receptor 2, blocked Ang II-dependent RhoGDI stabilization and proliferation at 12 and 24 h. In mice, Ang II infusion increased the intima-media thickness, collagen and myofiber production and VSMC proliferation, and these effects were shown to be dependent on RhoGDI1, RhoGDI2 and AT1 receptor. Ang II infusion exerted no significant effect on RhoGDI1 and RhoGDI2 protein levels, which were decreased after AT1 receptor inhibition. Conclusions: Together, the results of this study reveal a novel mechanism by which Ang II regulates RhoGDI stability by SUMOylation and ubiquitination via AT1 receptor activation and thus affects VSMC proliferation and vascular remodeling.
Article
Full-text available
Post-translational modification marked by the covalent attachment of the ubiquitin-like protein SUMO-1/ SMT3C has been implicated in a wide variety of cellular processes. Recently, two cDNAs encoding proteins related to SUMO-1 have been identified in human and mouse. The functions and regulation of these proteins, known as SUMO-2/SMT3A and SUMO-3/SMT3B, remain largely uncharacterized. We describe herein quantitative and qualitative distinctions between SUMO-1 and SUMO-2/3 in vertebrate cells, Much of this was accomplished through the application of an antibody that recognizes SUMO-2 and -3, but not SUMO-1. This antibody detected multiple SUMO-2/3-modified proteins and revealed that, together, SUMO-8 and -3 constitute a greater percentage of total cellular protein modification than does SUMO-1, Intriguingly, we found that there FT as a large pool of free, non-conjugated SUMO-2/3 and that the conjugation of SUMO-2/3 to high molecular mass proteins was induced when the cells were subjected to protein-damaging stimuli such as acute temperature fluctuation. In addition, we demonstrated that SUMO-2/3 conjugated poorly, if at all, to a major SUMO-1 substrate, the Ran GTPase-activating protein RanGAP1. Together, these results support the concept, of important distinctions between the SUMO-2/3 and SUMO-1 conjugation pathways and suggest a role for SUMO-2/3 in the cellular responses to environmental stress.
Article
Full-text available
DNA regulatory elements frequently harbor multiple recognition sites for several transcriptional activators. The response mounted from such compound response elements is often more pronounced than the simple sum of effects observed at single binding sites. The determinants of such transcriptional synergy and its control, however, are poorly understood. Through a genetic approach, we have uncovered a novel protein motif that limits the transcriptional synergy of multiple DNA-binding regulators. Disruption of these conserved synergy control motifs (SC motifs) selectively increases activity at compound, but not single, response elements. Although isolated SC motifs do not regulate transcription when tethered to DNA, their transfer to an activator lacking them is sufficient to impose limits on synergy. Mechanistic analysis of the two SC motifs found in the glucocorticoid receptor N-terminal region reveals that they function irrespective of the arrangement of the receptor binding sites or their distance from the transcription start site. Proper function, however, requires the receptor's ligand-binding domain and an engaged dimer interface. Notably, the motifs are not functional in yeast and do not alter the effect of p160 coactivators, suggesting that they require other nonconserved components to operate. Many activators across multiple classes harbor seemingly unrelated negative regulatory regions. The presence of SC motifs within them, however, suggests a common function and identifies SC motifs as critical elements of a general mechanism to modulate higher-order interactions among transcriptional regulators.
Article
Full-text available
SUMO-1 is a small ubiquitin-related modifier that is covalently linked to many cellular protein targets. Proteins modified by SUMO-1 and the SUMO-1-activating and -conjugating enzymes are located predominantly in the nucleus. Here we define a transferable sequence containing the ΨKXE motif, where Ψ represents a large hydrophobic amino acid, that confers the ability to be SUMO-1-modified on proteins to which it is linked. Whereas addition of short sequences from p53 and IκBα, containing the ΨKXE motif, to a carrier protein is sufficient for modification in vitro, modification in vivorequires the additional presence of a nuclear localization signal. Thus, protein substrates must be targeted to the nucleus to undergo SUMO-1 conjugation.
Article
Full-text available
Members of the protein inhibitor of activated signal transducer and activator of transcription (STAT) family (PIAS family) of proteins act as negative regulators of STATs in cytokine signaling. We report here that PIAS proteins have proapoptotic activity. PIAS1 induced apoptosis in both human 293T cells and human osteosarcoma U2OS cells. PIAS1 is localized in the nucleus as distinct nuclear dots. Ectopic expression of PIAS1 in U2OS cells activated JNK1 (c-Jun NH2-terminal kinase). A dominant-negative JNK1, capable of inhibiting PIAS1-induced JNK1 activation, blocked PIAS1-mediated apoptosis. Furthermore, a mutant PIAS1, lacking the first 9 amino acid residues, failed to repress Stat1-mediated gene activation although it retained its ability to activate JNK and to trigger apoptosis. Our results identify a novel function of PIAS1 in the induction of JNK-dependent apoptosis, independent of the previously known inhibitory activity of PIAS1 in STAT-mediated gene activation.
Article
Full-text available
MEK1, which is required for aggregation and chemotaxis in Dictyostelium, is rapidly and transiently SUMOylated in response to chemoattractant stimulation. SUMOylation is required for MEK1's function and its translocation from the nucleus to the cytosol and cortex, including the leading edge of chemotaxing cells. MEK1 in which the site of SUMOylation is mutated is retained in the nucleus and does not complement the mek1 null phenotype. Constitutively active MEK1 is cytosolic and is constitutively SUMOylated, whereas the corresponding nonactivatable MEK1 is not SUMOylated and nuclear. MEK1 is also ubiquitinated in response to signaling. A MEK1-interacting, ubiquitin E3 ligase RING domain-containing protein controls nuclear localization and MEK1 ubiquitination. These studies provide a pathway regulating the localization and function of MEK1.
Article
Full-text available
The RanGTPase activating protein RanGAP1 has essential functions in both nucleocytoplasmic transport and mitosis. In interphase, a significant fraction of vertebrate SUMO1-modified RanGAP1 forms a stable complex with the nucleoporin RanBP2/Nup358 at nuclear pore complexes. RanBP2 not only acts in the RanGTPase cycle but also is a SUMO1 E3 ligase. Here, we show that RanGAP1 is phosphorylated on residues T409, S428, and S442. Phosphorylation occurs before nuclear envelope breakdown and is maintained throughout mitosis. Nocodazole arrest leads to quantitative phosphorylation. The M-phase kinase cyclin B/Cdk1 phosphorylates RanGAP1 efficiently in vitro, and T409 phosphorylation correlates with nuclear accumulation of cyclin B1 in vivo. We find that phosphorylated RanGAP1 remains associated with RanBP2/Nup358 and the SUMO E2-conjugating enzyme Ubc9 in mitosis, hence mitotic phosphorylation may have functional consequences for the RanGTPase cycle and/or for RanBP2-dependent sumoylation.
Article
Activation of NF-κB is achieved by ubiquitination and proteasome-mediated degradation of IκBα. We have detected modified IκBα, conjugated to the small ubiquitin-like protein SUMO-1, which is resistant to signal-induced degradation. In the presence of an E1 SUMO-1-activating enzyme, Ubch9 conjugated SUMO-1 to IκBα primarily on K21, which is also utilized for ubiquitin modification. Thus, SUMO-1-modified IκBα cannot be ubiquitinated and is resistant to proteasome-mediated degradation. As a result, overexpression of SUMO-1 inhibits signal-induced activation of NF-κB-dependent transcription. Unlike ubiquitin modification, which requires phosphorylation of S32 and S36, SUMO-1 modification of IκBα is inhibited by phosphorylation. Thus, while ubiquitination targets proteins for rapid degradation, SUMO-1 modification acts antagonistically to generate proteins resistant to degradation.
Article
Post-translational modification marked by the covalent attachment of the ubiquitin-like protein SUMO-1/SMT3C has been implicated in a wide variety of cellular processes. Recently, two cDNAs encoding proteins related to SUMO-1 have been identified in human and mouse. The functions and regulation of these proteins, known as SUMO-2/SMT3A and SUMO-3/SMT3B, remain largely uncharacterized. We describe herein quantitative and qualitative distinctions between SUMO-1 and SUMO-2/3 in vertebrate cells. Much of this was accomplished through the application of an antibody that recognizes SUMO-2 and -3, but not SUMO-1. This antibody detected multiple SUMO-2/3-modified proteins and revealed that, together, SUMO-2 and -3 constitute a greater percentage of total cellular protein modification than does SUMO-1. Intriguingly, we found that there was a large pool of free, non-conjugated SUMO-2/3 and that the conjugation of SUMO-2/3 to high molecular mass proteins was induced when the cells were subjected to protein-damaging stimuli such as acute temperature fluctuation. In addition, we demonstrated that SUMO-2/3 conjugated poorly, if at all, to a major SUMO-1 substrate, the Ran GTPase-activating protein RanGAP1. Together, these results support the concept of important distinctions between the SUMO-2/3 and SUMO-1 conjugation pathways and suggest a role for SUMO-2/3 in the cellular responses to environmental stress.
Article
The RAD6 pathway is central to post-replicative DNA repair in eukaryotic cells; however, the machinery and its regulation remain poorly understood. Two principal elements of this pathway are the ubiquitin-conjugating enzymes RAD6 and the MMS2-UBC13 heterodimer, which are recruited to chromatin by the RING-finger proteins RAD18 and RAD5, respectively. Here we show that UBC9, a small ubiquitin-related modifier (SUMO)-conjugating enzyme, is also affiliated with this pathway and that proliferating cell nuclear antigen (PCNA) -- a DNA-polymerase sliding clamp involved in DNA synthesis and repair -- is a substrate. PCNA is mono-ubiquitinated through RAD6 and RAD18, modified by lysine-63-linked multi-ubiquitination--which additionally requires MMS2, UBC13 and RAD5--and is conjugated to SUMO by UBC9. All three modifications affect the same lysine residue of PCNA, suggesting that they label PCNA for alternative functions. We demonstrate that these modifications differentially affect resistance to DNA damage, and that damage-induced PCNA ubiquitination is elementary for DNA repair and occurs at the same conserved residue in yeast and humans.
Article
The ETS domain transcription factor Elk-1 is a direct target of the MAP kinase pathways. Phosphorylation of the Elk-1 transcriptional activation domain by MAP kinases triggers its activation. However, Elk-1 also contains two domains with repressive activities. One of these, the R motif, appears to function by suppressing the activity of the activation domain. Here, we demonstrate that SUMO modification of the R motif is required for this repressive activity. A dynamic interplay exists between the activating ERK MAP kinase pathway and the repressive SUMO pathway. ERK pathway activation leads to both phosphorylation of Elk-1 and loss of SUMO conjugation and, hence, to the loss of the repressive activity of the R motif. Thus, the reciprocal regulation of the activation and repressive activities are coupled by MAP kinase modification of Elk-1.