Vous êtes sur la page 1sur 8

Journal of Autoimmunity 84 (2017) 21e28

Contents lists available at ScienceDirect

Journal of Autoimmunity
journal homepage: www.elsevier.com/locate/jautimm

SUMO proteins: Guardians of immune system


Sabrina Adorisio a, Alessandra Fierabracci b, Isabella Muscari c, Anna Marina Liberati c,
Emira Ayroldi a, Graziella Migliorati a, Trinh Thi Thuy d, Carlo Riccardi a,
Domenico V. Delfino a, e, *
a
Section of Pharmacology, Department of Medicine, University of Perugia, Piazzale Severi, 06132, Perugia, Italy
b
Type 1 Diabetes Centre, Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Rome, Italy
c
Section of Onco-hematology, University of Perugia, Santa Maria Hospital, 05100, Terni, Italy
d
Institute of Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Ha Noi, Viet Nam
e
Foligno Nursing School, Via Oberdan 123, Foligno, PG, Italy

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

Article history: Small ubiquitin-like modifier (SUMO) proteins belong to the ubiquitin-like family and act to change the
Received 1 August 2017 function of target proteins through post-translational modifications. Through their interactions with innate
Received in revised form immune pathways, SUMOs promote an efficient immune response to pathogenic challenge avoiding, at the
4 September 2017
same time, an excess of immune response that could lead to the development of autoimmune diseases. This
Accepted 4 September 2017
Available online 15 September 2017
report discusses the general functions of SUMO proteins; highlights SUMO involvement in the innate immune
response through their role in NF-kB and interferon pathways; the involvement of SUMO proteins in auto-
immune diseases; and reviews bacterial, viral, and parasitic interactions with SUMO pathways. In conclusion,
Keywords:
Small ubiquitin-like modifier (SUMO)
we speculate that targeting SUMOs could represent a new therapeutic strategy against infections and
proteins autoimmunity.
Innate immune system © 2017 Elsevier Ltd. All rights reserved.
NF-kB pathway
Interferon pathway
Autoimmunity

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
2. Role of SUMO in innate immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
2.1. The NF-kB pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
2.2. The interferon pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
2.3. Crosstalk between NF-kB and IFN pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
3. Role of SUMO in autoimmunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
4. Role of SUMO during infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
4.1. Bacterial infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
4.2. Viral infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
4.2.1. The PML-nuclear bodies (NB) battle field . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
4.3. Parasitic infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
5. Role of SUMO in thymus function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
6. Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
6.1. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27

* Corresponding author. Section of Pharmacology, Department of Medicine, University of Perugia, Piazzale Severi, 06132, Perugia, Italy.
E-mail address: domenico.delfino@unipg.it (D.V. Delfino).

http://dx.doi.org/10.1016/j.jaut.2017.09.001
0896-8411/© 2017 Elsevier Ltd. All rights reserved.
22 S. Adorisio et al. / Journal of Autoimmunity 84 (2017) 21e28

1. Introduction pathogens and dissect how SUMO modulation impacts the immune
response to infection and autoimmunity.
Post-translational modifications by small ubiquitin-like modi-
fiers (SUMOs) are crucial in activating protein functions, which lead 2. Role of SUMO in innate immunity
to target effects at the molecular, cellular, and systemic levels.
SUMOs belong to the ubiquitin-like (Ubl) family of proteins and are The innate immune response forms the first line of defense
ubiquitously expressed in eukaryotic cells. Four SUMO isoforms against invading pathogens. One important aspect of innate im-
have been identified, SUMO-1-4, with SUMO-2 and -3 almost munity is the recognition of pathogens and microorganisms that
identical in structure [1]. Conjugation of SUMO proteins with target express pathogen-associated molecular patterns (PAMPs) by host
substrates, sumoylation, resembles the ubiquitination process and cell pattern recognition receptors (PRRs). Activation of these re-
involves three distinct steps. First, SUMO binds to the E1 activating ceptors triggers signaling pathways that result in the production of
enzyme, SAE1/SAE2, which results in the activation of SUMO. anti-microbial mediators and the initiation of an immune response.
Activated SUMO is then transferred to the E2 conjugation enzyme, PRR signaling cascades, stemming from both membrane-bound
Ubc9. Ubc9 then acts to target SUMO to its specific substrate, which and intracellular receptors, culminate in the activation of the
is bound to an E3 ligase (protein inhibitor of activated STAT [PIAS], transcriptional regulators, nuclear factor kappa-light-chain-
Ran binding protein 2 [RanBP2], polycomb protein 2 [Pc2]), through enhancer of activated B cells (NF-kB) or interferon regulatory fac-
covalent binding. Sumoylation differs from ubiquitination in the tors (IRF) [23].
number of enzymes involved, with less SUMO-related enzymes These pathways are tightly regulated by sumoylation [24,25],
known, and that E3 ligation is not always required for SUMO which promotes the production of anti-microbial molecules. SUMO
binding. In the consensus site on the target protein, characterized modifications are also important in the negative regulation of these
by the “JKXE” motif [2], SUMO binds to a lysine and then is pathways, avoiding an excessive immune response that could be
released from the target protein through desumoylation, catalyzed detrimental to the host and lead to the development of autoim-
by SUMO-specific peptidases (SENP1, 2, 3, 5, 6, 7) [3]. Thus, mune disease or tissue damage. This balance between activation
sumoylation is a dynamic and reversible post-translational modi- and resolution of inflammatory cascades is exemplified in the
fication process [4]. cellular response to viral infection. Viral DNA is recognized by cyclic
Although SUMO proteins have no function on their own, they GMP-AMP synthase (cGAS), which results in the activation of the
are capable of modulating the functionality of hundreds of target adaptor protein, stimulator of interferon genes (STING). Tripartite
proteins, with effects that are diverse and target protein-specific. motif containing 38 (TRIM38), a ubiquitin ligase, sumoylates and
Recently, SUMOs' role in the regulation of DNA activities, such as stabilizes cGAS and STING during the early phase of viral infection,
DNA synthesis and damage repair, has emerged as crucial. For amplifying the antiviral cell response. In the late phase of infection,
example, SUMO regulates double-stranded break repair, a process cGAS and STING are desumoylated by sentrin-specific protease 2
critically important for maintenance of genome stability [4]. SUMO (Senp2) and subsequently degraded, thus shutting off this pathway
is also involved in DNA-protein crosslink repair in yeast [5], binding and limiting the immune response to avoid excess activation
to proliferating cell nuclear antigen (PCNA) and signaling the [26,27].
recruitment of the anti-recombinogenic DNA helicase, Srs2 [6]. In
addition, sumoylation of the phosphatase and tensin homolog 2.1. The NF-kB pathway
protein (PTEN) is required for the PTEN-dependent DNA damage
response [7]. Among PRRs, Toll-like receptors (TLRs) form the frontline of
Sumoylation can occur at multiple points in the same pathway. immune defense. Upon ligand binding to membrane-bound TLRs, a
Notably, during meiotic homologous recombination, there is a signaling cascade is initiated that results in NF-kB activation, the
concomitant sumoylation of many proteins, which are then sub- canonical inflammatory transcription factor (Fig. 1). For example,
sequently desumoylated to shut down the pathway [8]. In partic- when TLR4 recognizes lipopolysaccharide (LPS) on bacterial outer
ular, the interference phenomenon of meiotic crossover is membranes, this triggers the recruitment of the adaptor protein
regulated by sumoylation, where topoisomerase II and Red1 myeloid differentiation primary response gene 88 (MyD88) and
sumoylation is a critical control mechanism [9]. Additionally, Ubc9 signaling molecules interleukin-1 receptor-associated kinases 1
sumoylation determines the SUMO-modification state of target and 4 (IRAK1, 4). IRAK1 and IRAK4 then associate with TNF
proteins, where switching from mono-sumoylation to poly- receptor-associated factor 6 (TRAF6). TRAF6 activates the down-
sumoylation, results in a change of function, essential for meiotic stream TAK kinase, which leads to inhibitor of nuclear factor kappa-
cell division [10,11]. Sumoylation events also safeguard somatic cell B kinase (IKK) complex activation. IKK complex then regulates the
identity through repressing transition from one cell type to a activation state of NF-kB, leading to nuclear translocation and the
different cell type [12]. transcription of inflammatory cytokines and initiation of the innate
Sumoylation regulates many other processes that are important immune response [1].
for cellular function. SUMO influences gene transcription, through In this pathway, discussed further below, SUMOs act at different
both activation [13] and repression [14]. Intermediate filament levels to influence signaling. At the beginning of the cascade, the
proteins are also modified by SUMO, which are important for Pellino protein associates with IRAK1, resulting in reciprocal
cytoskeleton structural functions and intracellular communications phosphorylation. Pellino-1 is modified by SUMO-1, although the
[15]. SUMO-2 plays a role in repressing the expression of provirus biological significance of this modification is still unclear [28].
and endogenous retroviruses by pluripotent stem cells [16]. Downstream of IRAK-1/TRAF6, SUMO-1 binds to TRAF family
SUMO modifications have also been implicated in general health member-associated NF-kappa-B activator (TANK) and relieves its
maintenance and systemic disease. Recent discoveries suggest that repression on TLR signaling. Acting on the IKK complex, SUMO-3
sumoylation plays a role in breast cancer [17], melanoma [18], renal binds to IKK-g (NEMO) which forms a complex with IKKa and
carcinoma [19], and the response to heart failure therapy [20e22]. IKKb kinases, that then phosphorylate IkBa, an inhibitor of NF-kB,
Given these wide-ranging effects and the importance of SUMO- leading to its degradation and subsequent NF-kB activation.
dependent processes on health, the remainder of this review will SUMO-1 also negatively regulates this pathway through binding
examine the role of SUMO in the innate immune defense against and protecting dephosphorylated IkBa from degradation, thus
S. Adorisio et al. / Journal of Autoimmunity 84 (2017) 21e28 23

mechanism involved in innate immunity [30,31].

2.2. The interferon pathway

Viruses stimulate an innate immune response by triggering the


production of type I and II interferons (IFNs). IFNs bind to their
receptors, IFN alpha receptor (IFNAR) and IFN gamma receptor
(IFNGR), initiating signal transduction pathways that lead to tran-
scription of interferon-stimulated genes (ISGs) and IFN gamma-
activated site (GAS) genes, which contribute to the antiviral im-
mune response. These pathways are rigorously regulated by
sumoylation at multiple sites. Viral products, such as CpG-rich DNA
and single-stranded (ss)RNA, trigger signaling cascades that lead to
IRF3, IRF7, and IRF8 transcription factor activation, which then
stimulate the transcription of ISGs (Fig. 2). IRF3 sumoylation in-
hibits the IRF3-dependent transcription of IFNb, whereas desu-
moylation, by SENP2, results in ubiquitination and proteasomal
degradation of IRF3, thus removing the IRF3-SUMO-dependent
inhibition of IFN-b transcription [33]. IRF3 is also activated by
sumoylated promyelocytic leukemia protein (PML)-IV. Sumoylated
PML-IV associates with peptidyl-prolyl cis/trans isomerase (Pin1),
preventing Pin1 from binding and degrading IRF3. This leads to an
increased production of IRF3-dependent IFN-b and a resulting
strong antiviral response [34,35]. Therefore, SUMO both promotes
and inhibits antiviral IRF3 activity, through direct binding (inhibi-
tion) and PML-IV sumoylation (promotion) that impedes the Pin1-
dependent degradation of IRF3 (Fig. 2).
IRF7 is phosphorylated and activated through MyD88-
dependent TLR activation, which occurs when intracellular TLR9
and TLR7/8 recognize CpG DNA and ssRNA viral products (Fig. 2).
SUMO binds and inhibits IRF7 phosphorylation, resulting in

Fig. 1. The role of SUMOylation in the NF-kB signaling pathway. SUMOylation and
deSUMOylation of proteins are observed at several steps in this pathway and can both
positively (green) and negatively (red) regulate NF-kB activation.

keeping NF-kB in an inactive state. The detachment of SUMO from


IkBa results in NF-kB activation. In another example of negative NF-
kB regulation, DNA damage induces the desumoylation of NEMO by
SUMO-specific protease 2 (SENP2), resulting in inhibition of NF-kB
activation [29]. We speculate that this mechanism of NF-kB
repression may also occur in response to innate immune signaling
(Fig. 1). Therefore, SUMO proteins play a dual role in the NF-kB
pathway: activation, through the sumoylation of TANK and NEMO,
to mount an efficient innate immune response; and inhibition,
through sumoylation of IkBa, in order to limit excessive activation
[1]. This pathway regulation by SUMO is extremely important.
Either deficient or excessive NF-kB activation may be detrimental to
the host's response against pathogens, leading to a decreased
ability to defend against infection or chronic inflammation that
may promote the development of autoimmune diseases.
NF-kB transcriptional activity is also regulated through inter-
action with nuclear co-repressor (NCoR) complex. TLR signaling
results in the release of NCoR from its DNA binding site, allowing
access for NF-kB binding. However, liver X receptor (LXR) sumoy-
lation, mediated by the E3 SUMO ligase, PIAS1, maintains NCoR
binding, thus repressing NF-kB-mediated transcription [30,31]. The
TLR-dependent de-repression of inflammatory target genes
through NCoR release relies on both the action of coronin 2A
(CORO2A), a component of the NCoR complex, and LXR desumoy-
lation by SENP3. Sumoylated LXRs bind and inhibit CORO2A [32] Fig. 2. The role of SUMOylation in the interferon pathway. SUMOylation and
deSUMOylation of IRF transcription factors can inhibit translation and phosphorylation
(Fig. 1). This regulatory mechanism is involved in the promotion or trigger degradation, respectively. Sumoylation of upstream factors can both activate
of inflammation in atherosclerosis but may also be a general (green) and inhibit (red) IRF transcription factors.
24 S. Adorisio et al. / Journal of Autoimmunity 84 (2017) 21e28

negative regulation of IFN-a expression [1]. Enhancing this complex occurs, it completely blocks the IFN-g pathway through
repression, TRIM28 functions as a SUMO E3 enzyme, which inhibiting phosphorylation of the sumoylated homodimer (Fig. 3).
sumoylates IRF7, resulting in transcriptional inhibition of the IFN-a SUMO-1 also binds to PML, however, this sumoylation does not
gene [36]. In addition to TRIM28, another SUMO E3 enzyme, protein reduce its STAT1 phosphorylation activity. Similar to SUMO-1,
inhibitor of activated STAT-1 (PIAS), inhibits IFN transcription SUMO-3 also modifies STAT1. However, SUMO-3 binding to PML
through SUMO modifications [37] (Fig. 2). IRF8, when bound to results in inhibition rather than reinforcement of STAT1 phos-
SUMO-2/3, represses the transcription of its antiviral target genes phorylation [38]. Therefore, SUMO-1 and SUMO-3 modifications of
interleukin (IL)-12 p40 and IFN. Transcription of these genes are STAT1 and PML impair ISG transcription and the IFN-g-mediated
then activated by SENP1-dependent desumoylation of IRF8 [35] antiviral response, while they do not affect these outcomes for type
(Fig. 2). I IFNs.
Following production and release, type I and type II IFNs bind to IFN-g-induced gene transcription is also controlled by LXRs. LXR
their receptors on the cell membrane, IFNAR and IFNGR, respec- sumoylation negatively regulates the transcription of the nitric
tively, and initiate signaling pathways that result in ISG gene oxide synthase 2 (NOS) gene, induced by IFN-g, as it decreases
transcription and enhancement of the antiviral immune response. STAT1 recruitment to the gene promoter. Of note, the inhibitory
Specifically, type I IFNs (a and b), bind IFNAR and stimulate the effect is bidirectional, as IFN-g inhibits LXR-dependent up-regula-
recruitment of Janus kinase 1 (JAK1) and tyrosine-protein kinase 2 tion of selective gene targets such as ATP-binding cassette A1
(TYK2), which, in turn, phosphorylate STAT1/STAT2 heterodimers (ABCA1) and sterol response element binding protein 1c (SRBP1c)
that form a complex with IRF9 (Fig. 3). This trimeric complex [39].
translocates to the nucleus where it binds to DNA consensus se-
quences, interferon-sensitive response elements (ISRE), promoting 2.3. Crosstalk between NF-kB and IFN pathways
the transcription of ISGs genes. Similarly, when IFN-g, a type II IFN,
binds to its receptor, IFNGR, JAK1/JAK2 are recruited to the receptor SUMO has been implicated in providing balance to the antiviral
and phosphorylate the STAT1 homodimer. This complex trans- response by interfering with both IRF3- and NF-kB-dependent
locates to the nucleus and binds to the DNA consensus sequence production of type I IFNs and cytokines. During a viral infection,
GAS to stimulate the transcription of ISG genes. PML promotes this viral DNA binds to the cytosolic PRR, retinoic acid-inducible gene-I
pathway by increasing the phosphorylation of STAT1; however, (RIG-I), which is sumoylated [40] and activates the mitochondrial
SUMO-1 binds and inhibits STAT1 phosphorylation. antiviral signaling (MAVS) protein. MAVS then stimulates the NF-kB
Whereas STAT1 sumoylation does not affect the type I IFN and IRF3-dependent production of type I IFNs. This antiviral
pathway, as alternative phosphorylation of the STAT2 homodimer response is inhibited by MAVS degradation, which is promoted by
insulin receptor tyrosine kinase substrate (IRTKS)-dependent
sumoylation of poly(rC)-binding protein 2 (PCBP2). Sumoylated
PCBP2 is exported from the nucleus to the cytoplasm where it de-
grades MAVS [34].

3. Role of SUMO in autoimmunity

The importance of SUMO proteins in avoiding excessive immune


response and subsequent autoimmunity is demonstrated by their
involvement in both the regulation of T regulatory (Treg) cell ac-
tivity and the development of different autoimmune diseases. In
the case of CD4þCD25þFoxp3þ Treg cells, selective deletion of Ubc9,
the E2 SUMO-conjugation enzyme, in Tregs results in fatal, early-
onset autoimmunity similar to that seen in Foxp3-mutant mice
[41].
SUMO has been shown to play a role in several autoimmune
diseases, including type 1 diabetes (T1D) [42e47], Bechet's disease
[48], rheumatoid arthritis (RA) [49], gammopathies [50], and
Wiskott-Aldrich syndrome (WAS) [51] (Table 1). T1D is a multi-
factorial autoimmune disease that leads to the destruction of
pancreatic insulin-producing b cells [47]. SUMO4 could play a
central role in T1D. It was initially cloned from the IDDM5 region of
chromosome 6q25 [42], specifically among 55 single-nucleotide
polymorphism markers located in the 16 non-HLA genes geno-
typed to T1D [43]. SUMO4 is known to interact with IkBa and
inhibit NF-kB transcriptional activity. The SUMO4 M55V mutation
prevents the sumoylation of IkBa and subsequent inhibition of NF-
kB. Downstream, this mutation leads to the activation and tran-
scription of genes involved in T1D development [44,45]. Thus, NF-
kB dysregulation through a SUMO4-mediated mechanism could be
an important factor in the pathogenesis of T1D [44e46].
SUMO4 also is involved in HLA-B51-positive Bechet's disease.
The C438T polymorphism of SUMO4 is associated with a signifi-
Fig. 3. The role of SUMOylation in interferon-activated JAK/STAT pathways.
cantly increased risk of papulopustular skin lesions [48]. In RA, the
SUMOylation of STAT1 can impair and block gene expression activated by type I and SUMO-conjugating enzyme UBC9 may promote the proliferation
type II interferons, respectively. and migration of fibroblast-like synoviocytes in affected joints [49].
S. Adorisio et al. / Journal of Autoimmunity 84 (2017) 21e28 25

Table 1 process in host cells. Here, we will review the literature on SUMO
SUMO involvement in autoimmune and infectious diseases. modifications during bacterial, viral, and parasitic infections
Diseases Pathogens References (Table 1).
Autoimmunity
Type 1 diabetes 42, 43, 44, 45, 46, 47 4.1. Bacterial infection
Bechet's disease 48
Rheumatoid arthritis 49 A landmark finding demonstrating the importance of sumoy-
Gammopathies 50
lation during the host response to bacterial infection, was the dis-
Wiskott-Aldrich syndrome 51
Breast cancer 52 covery of the evasion strategy of Listeria monocytogenes, which
Infections results in a reduction in host cellular sumoylated proteins critical
Bacteria L. monocytogenes 54, 55 for defense against infection, mediated by proteasome-
Shigella 56, 57
independent Ubc9 degradation by the bacterial virulence factor
Xanthomonas euvesicatoria 58, 59
Pseudomonas aeruginosa 60
listeriolysin O (LLO). In vitro studies show that overexpression of
Viruses Kaposi's sarcoma-associated 65, 68, 79, 80, 81 SUMO1 or SUMO2 decreases the number of L. monocytogenes in
herpes virus infected HeLa cells. This demonstrates the importance of sumoy-
Vaccinia virus 66 lation in the resistance of host cells to infective bacteria, as it is
Enterovirus 71 67, 68, 69
detrimental for bacterial invasion and replication [54,55].
Epstein-Barr virus 68, 69, 70, 76
Crimean-Congo 71 Similar to listeria infection, Shigella also impairs the sumoyla-
hemorrhagic fever virus tion process in infected cells. Specifically, like L. monocytogenes,
Vesicular stomatitis virus 72, 73 Shigella dramatically decreases the amount of Ubc9, an E2 conju-
Herpes simplex type 1 virus 75, 82 gating enzyme, and therefore, decreases the amount of sumoylated
Cytomegalovirus 77
Adenovirus 78
proteins important in the host defense against infection. This effect
Parasites Trypanosoma brucei 83 is mediated by the type III secretion system (T3SS), a system that
Shigella uses to deliver effector proteins into the cytosol of infected
human cells [56]. One such effector is OspF, which enters the nu-
In addition, the presence of heat-shock protein 90 (HSP90)-SUMO1 cleus and regulates pro-inflammatory cytokine expression. OspF
is a marker of healthy individuals at risk for plasma cell dyscrasias. translocation into the nucleus is dependent on its sumoylation [56].
Further, the dominant inheritance of this posttranslationally This discovery has important public health implications, as
modified autoantigenic paratarg is one of the strongest molecular- Shigella-dependent diarrheal disease is a major health problem in
level risk factors for developing monoclonal gammopathies of un- the developing world. Increasing protein sumoylation is a potential
determined significance (MGUS), multiple myeloma (MM), and therapeutic strategy against Shigella infection, as Shigella is
Waldenstrom's macroglobulinemia (WM) [50]. becoming increasingly resistant to antibiotic treatment [57].
The role of SUMO in WAS highlights the balancing act between Another interesting bacterial strategy that interferes with
immune and autoimmune response played by SUMO. WAS is an X- sumoylation is described in plants. In tomato plants, Xanthomonas
linked disorder that causes both a deficient immune response and euvescicatoria (Xcv) produces XopD, a type III secretion effector,
autoimmunity. It is caused by mutations in WAS, which encodes that desumoylates the tomato transcription factor SIERF4, thus
Wiskott-Aldrich syndrome protein (WASp). In patients with WAS, repressing the tomato's anti-Xcv immunity, which relies on
autoimmunity is favored over immune response when the SUMO- sumoylated SIERF4 [58,59].
binding site of WASp bears the V75M mutation, one of many In a study analyzing host interactions with Pseudomonas aeru-
disease-causing mutations. This specific mutation compromises ginosa and Streptococcus pneumonia, two of the most common
WASp sumoylation, causing histone deacetylase-6 (HDAC6) to bacteria that cause pneumonia, SUMO1 has been shown to be
upregulate the NF-kB response genes associated with autoimmu- important in defense during infection [60]. This is another example
nity (granulocyte macrophage colony-stimulating factor [GM-CSF], of SUMO proteins that are good therapeutic targets. Like Shigella,
tumor necrosis factor alpha-induced protein 2 [TNFAIP2], IL-1b) but pathogens that cause pneumonia are becoming increasingly resis-
not adaptive immunity (IFNG, STAT1, TLR1) in Th1 cells. Moreover, tance to antibiotics and therefore, new drugs and strategies are
expression of sumoylation-deficient WASp leads to ectopic devel- needed to combat disease [60].
opment of the TH17-like phenotype, which favors the development
of WAS-associated autoimmune manifestations [51]. 4.2. Viral infection
SUMO-related autoimmunity may contribute to certain cancers.
The presence of autoantibodies to SUMO peptidase 2 and other As seen in bacteria, viruses also interfere with SUMO pathways
important centrosome antigens have been shown to be associated as a strategy to evade the host immune system. Many viruses have
with breast cancer, suggesting that alterations in sumoylation could been identified which disrupt SUMO modifications (Table 1). These
be related to the development of autoimmunity in breast cancer include the chick embryo lethal orphan (CELO) virus, geminivirus,
patients [52]. SUMOs also are important regulators of autoimmu- adenovirus, Kaposi's sarcoma-associated herpes virus (KSHV),
nity in plants. For example, the SUMO E3 ligase siz1 regulates vaccinia virus (VACV), bovine papillomavirus, human papilloma-
numerous processes, including immune response, in Arabidopsis. A virus, Moloney murine leukemia virus, influenza A virus, cyto-
loss-of-function mutation in siz1 produces an autoimmune megalovirus, Epstein-Barr virus (EBV), Hantaan virus, Mason-Pfizer
phenotype [53]. Thus, SUMO proteins are critical molecules that virus, human immunodeficiency virus, Ebola Zaire virus, herpes
can control autoimmunity development. simplex type 1 virus, varicella zoster virus, and encephalomyocar-
ditis virus [61] and others, described in more detail below (see
4. Role of SUMO during infection Table 1). Viruses disrupt sumoylation through two established
ways: 1) various viral proteins are sumoylated to exploit the host
The importance of sumoylation in the immune response during SUMO machinery; and 2) they interfere with the sumoylation of
infection is highlighted by the development of pathogen strategies host immune defense proteins, through desumoylation (targeting
that aim to evade host defense by impairing the sumoylation SUMOs and sumoylated proteins for ubiquitin-mediated
26 S. Adorisio et al. / Journal of Autoimmunity 84 (2017) 21e28

degradation) or sumoylation. The ultimate goal of both strategies is consequence, Sp100A, within PML-NBs, create a permissive envi-
to decrease the host innate immune response [62e64]. ronment for viral replication [78]. In addition, KSHV encodes
KSHV employs the first strategy to produce a transcriptional SUMO-targeting E3 ubiquitin ligase (STUbL), K-Rta, which targets
repressor called K-bZIP, which interferes with the type I IFN sumoylated PML involved in virus replication inhibition for
pathway when sumoylated, without E3-SUMO ligase activity [65]. ubiquitin-mediated degradation [68,79].
VACV exploits the host cell SUMO conjugation system to induce a KSHV utilize at least other two proteins to dampen the SUMO-
covalent interaction between the E3 protein and SUMO1 or SUMO2. dependent host immune response. Viral (v)IRF3 promotes the
This results in modifications that repress E3 transcriptional regu- SUMO-dependent ubiquitination and degradation of PML and vPK
lation of p53-upregulated modulator of apoptosis (PUMA) and catalyzes the phosphorylation-dependent sumoylation of the
apoptotic protease activating factor 1 (APAF-1) genes [66]. More- transcriptional repressors K-bZIP and KAP-1. Additional sumoylated
over, during enterovirus 71 infection, polymerase 3D sumoylation proteins are also involved in the KSHV evasion of host defense
facilitates virus replication and infection, leading to fatal neuro- mechanisms and all of them, including ORF7, interfere with the
logical diseases in children [67e69]. assembly of PML-NBs [80]. KSHV also encodes latency-associated
With regard to the second strategy, various viruses acquire the nuclear antigen (LANA), a multifunctional protein that interacts
ability to sumoylate host proteins. EBV latent membrane protein 1 with chromatin factors. LANA displaces Sp100 from chromatin and
(LMP1) sumoylates IRF7, limiting its function in initiating an effi- relocalizes it in PML-NBs by enhancing SUMO-1 and SUMO-2
cient innate immune response [68e70]. In hepatocytes infected binding [81].
with Crimean-Congo hemorrhagic fever virus (CCHFV), there is an PML-NBs are also the targets of herpes simplex virus 1 (HSV-1),
up-regulation of SUMO-E1 enzyme (SAE2/UBA2), although the although by a different mechanism. HSV-1 encodes ICP0, an E3
exact role of sumoylation remains to be clarified [71]. In addition, ubiquitin ligase, that targets proteins for ubiquitination and
viruses, such as vesicular stomatitis virus (VSV), promote them- consequent degradation, which is required for efficient viral repli-
selves through inducing p53 sumoylation. This modification leads cation. In the PML-NB, ICP0 desumoylates PML-NB proteins
to cellular senescence, which favors viral replication [72]. involved in repression, decreasing the overall level of SUMO-
Host cells also use sumoylation to regulate and activate the conjugated proteins in the infected cells and allowing a permis-
immune response against infection. One example is the increased sive environment for viral replication [82].
resistance of human cells to VSV infection due to SUMO-dependent
degradation protection of the human MxA protein, a member of the 4.3. Parasitic infection
large dynamin-like GTPase family. MxA is stabilized by SUMO and
inhibits VSV primary transcription. This effect is VSV-specific as Parasites have an original mechanism to avoid the host immune
SUMO actually renders cells more sensitive to the rabies virus response by exploiting SUMO. Trypanosoma brucei displays the
(RABV) [73]. variant surface glycoprotein (VSG) on its surface, which allows the
It is interesting to examine the differential role of SUMO-1 and parasite to evade detection by extensive antigenic variation. Try-
SUMO-2/3 during viral reactivation. Chromatin occupancy of panosoma brucei express only one VSG variant among approxi-
SUMO-2/3, but not of SUMO-1, is increased during viral reactivation mately 1000 VSG genes, allowing it to avoid the host immune
in transcription factor binding regions. This suggests a role for antibody response. This monoallelic expression of VSG is a result of
SUMO-2/3 in host-mediated suppression of gene expression [74]. In SUMO, which associates with the VSG chromatin region and the
another study, a comprehensive analysis of cellular proteins whose nuclear body ESB. Sumoylation of chromatin-associated proteins
sumoylation state was altered during HSV1 infection showed that determines the recruitment of RNA polymerase I to the VSG pro-
sumoylation was reduced in 124 proteins. Among these, several moter. This suggests that sumoylation is involved in VSG mono-
proteins were targets of infected cell protein 0 (ICP0), which allelic expression [83].
functions as an E3 ubiquitin ligase, targeting proteins for ubiquiti-
nation and degradation with putative repressive activity [75]. 5. Role of SUMO in thymus function
A fascinating emerging strategy employed by viruses to evade
the host immune response is through coding microRNAs (miRNAs). Along with the bone marrow, the thymus is the primary
A recent study identified miRNAs encoded by EBV that interfere lymphoid organ where T lymphocyte maturation is completed.
with the SUMO signaling network of the host, that influence the Therefore, the thymus plays an important role in the defense
immune response, such as PML, IRF1, Sp100, HLA-B, MX-1, ADAR, against pathogens. During infections, the thymus undergoes
OAS3, RNF4, and STAT6 [76]. reversible involution [84]. Glucocorticoids (GCs) are involved in
thymic involution by triggering massive thymocyte apoptosis. To
4.2.1. The PML-nuclear bodies (NB) battle field this end, GCs stimulate the transcription of glucocorticoid-induced
An exemplification of the fight between viruses and host cells, in leucine zipper (Gilz) and the activation of caspase-8, which, in turn,
which SUMO has a starring role, is the viral interference with co-activate each other. Gilz activates caspase-8 and activated
protein sumoylation in the context of PML-NB. Recently it has been caspase-8 sumoylates Gilz, allowing the prolongation of Gilz
shown, that host cells have the ability to counterattack viruses by expression by inhibiting ubiquitin-mediated Gilz proteasomal
sumoylating viral proteins within PML-NBs. For example, the ND10 degradation. The final result of this process is apoptosis of thy-
component of PML proteins sumoylates the major immediate early mocytes [85,86].
protein IE1p72 of human cytomegalovirus, thus compromising its
interferon-antagonistic function [77]. Different PML-NBs- 6. Discussion
associated proteins are sumoylated by SUMO-1, within the
external NB formed by PML and Sp100, and SUMO-2/3, within the SUMO molecules bind to target proteins and modulate their
internal NB. Adenovirus disrupt this antiviral mechanism through function. The goal of this review was to highlight how SUMO plays a
transactivator protein E1A, which targets PML-II and promotes viral crucial role in innate immunity, reviewing the latest findings on
and cellular transcription. Adenovirus also induce the detachment SUMO's influence during the innate immune response and
of SUMO-2/3 from Sp100A, thus impeding its association with infection.
heterochromatin protein 1 (HP1), a repressive factor. As a From the findings reported, three important points emerged: 1)
S. Adorisio et al. / Journal of Autoimmunity 84 (2017) 21e28 27

SUMO proteins act at multiple sites in a pathway and often, the Cell Biol. 17 (2016) 379e394.
[5] J. Stingele, M.S. Schwarz, N. Bloemeke, P.G. Wolf, S. Jentsch, A DNA-dependent
sumoylation of all these sites is necessary for the function of that
protease involved in DNA-protein crosslink repair, Cell 158 (2014) 327e338.
particular pathway; 2) SUMOs are able to enhance signaling that [6] A.A. Armstrong, F. Mohideen, C.D. Lima, Recognition of SUMO-modified PCNA
initiates an immune response, but also are important in resolving requires tandem receptor motifs in Srs2, Nature 483 (2012) 59e63.
inflammation, allowing a balanced response; 3) SUMOs are [7] C. Bassi, J. Ho, T. Srikumar, R.J. Dowling, C. Gorrini, S.J. Miller, et al., Nuclear
PTEN controls DNA repair and sensitivity to genotoxic stress, Science 341
important molecules in defense against infections, as pathogens (2013) 395e399.
interfere with sumoylation of host molecules to evade an immune [8] I. Psakhye, S. Jentsch, Protein group modification and synergy in the SUMO
response. pathway as exemplified in DNA repair, Cell 151 (2012) 807e820.
[9] L. Zhang, S. Wang, S. Yin, S. Hong, K.P. Kim, N. Kleckner, Topoisomerase II
During an innate immune response, SUMO proteins influence mediates meiotic crossover interference, Nature 511 (2014) 551e556.
both the NF-kB and IFNs pathways, the two major innate immune [10] M. Dasso, Biochemistry: rear view of an enzyme, Nature 497 (2013) 576e577.
pathways, at multiple levels. As has been shown for SUMO- [11] H. Klug, M. Xaver, V.K. Chaugule, S. Koidl, G. Mittler, F. Klein, et al., Ubc9
sumoylation controls SUMO chain formation and meiotic synapsis in
dependent homologous recombination, concurrent SUMO modifi- Saccharomyces cerevisiae, Mol. Cell 50 (2013) 625e636.
cations are necessary for the proper functioning of each immune [12] S. Cheloufi, U. Elling, B. Hopfgartner, Y.L. Jung, J. Murn, M. Ninova, et al., The
pathway. In addition, SUMO function during an immune response histone chaperone CAF-1 safeguards somatic cell identity, Nature 528 (2015)
218e224.
varies, being important in both increasing the activation of innate [13] L. Yang, C. Lin, W. Liu, J. Zhang, K.A. Ohgi, J.D. Grinstein, et al., ncRNA- and Pc2
immune signaling pathways through sumoylation, and decreasing methylation-dependent gene relocation between nuclear structures mediates
the innate immune response through desumoylation or binding to gene activation programs, Cell 147 (2011) 773e788.
[14] P.A. Dutchak, T. Katafuchi, A.L. Bookout, J.H. Choi, R.T. Yu, D.J. Mangelsdorf, et
inhibitory proteins. This dual-action contributes to a balanced and
al., Fibroblast growth factor-21 regulates PPARgamma activity and the anti-
well-regulated immune response that is capable of protection diabetic actions of thiazolidinediones, Cell 148 (2012) 556e567.
against foreign pathogens, but at the same time, prevented from an [15] N.T. Snider, M.B. Omary, Post-translational modifications of intermediate
exaggerated response, involving the risk of autoimmunity and tis- filament proteins: mechanisms and functions, Nat. Rev. Mol. Cell Biol. 15
(2014) 163e177.
sue damage. Type 1 diabetes development is, in fact, partly a [16] B.X. Yang, C.A. El Farran, H.C. Guo, T. Yu, H.T. Fang, H.F. Wang, et al., Systematic
consequence of SUMO-NEMO-dependent augmentation of NF-kB identification of factors for provirus silencing in embryonic stem cells, Cell
activity [46]. Thus, the SUMO system must strictly control immune 163 (2015) 230e245.
[17] J.D. Kessler, K.T. Kahle, T. Sun, K.L. Meerbrey, M.R. Schlabach, E.M. Schmitt, et
signaling pathway activation. al., A SUMOylation-dependent transcriptional subprogram is required for
Myc-driven tumorigenesis, Science 335 (2012) 348e353.
[18] S. Yokoyama, S.L. Woods, G.M. Boyle, L.G. Aoude, S. MacGregor, V. Zismann, et
6.1. Conclusions
al., A novel recurrent mutation in MITF predisposes to familial and sporadic
melanoma, Nature 480 (2011) 99e103.
The importance of sumoylation in autoimmunity is highlighted [19] C. Bertolotto, F. Lesueur, S. Giuliano, T. Strub, M. de Lichy, K. Bille, et al.,
by the involvement of SUMO proteins in the development of A SUMOylation-defective MITF germline mutation predisposes to melanoma
and renal carcinoma, Nature 480 (2011) 94e98.
different autoimmune diseases and its importance in defense [20] C. Kho, A. Lee, D. Jeong, J.G. Oh, A.H. Chaanine, E. Kizana, et al., SUMO1-
against pathogens. For example, bacteria, viruses, and parasites dependent modulation of SERCA2a in heart failure, Nature 477 (2011)
interfere with the sumoylation of host cells by exploiting the host 601e605.
[21] S.K. Shenoy, H.A. Rockman, Cardiovascular biology: heart fails without pump
system to enable sumoylation of their own proteins and by inter- partner, Nature 477 (2011) 546e547.
fering with sumoylation of host proteins. Both strategies lead to a [22] J.J. McMurray, G.L. Smith, Calcium handling in the failing heart and
decreased immune defense or detection, which increasing their SUMOeweighing the evidence, N. Engl. J. Med. 365 (2011) 1738e1739.
[23] K. Chen, J. Liu, X. Cao, Regulation of type I interferon signaling in immunity
infective potential. The pivotal role of SUMO in modulation of the and inflammation: a comprehensive review, J. Autoimmun. 83 (2017) 1e11.
immune system suggests that new therapeutic strategies for [24] Z. Hannoun, G. Maarifi, M.K. Chelbi-Alix, The implication of SUMO in intrinsic
treating autoimmune diseases and infections that target SUMO are and innate immunity, Cytokine & Growth Factor Rev. 29 (2016) 3e16.
[25] J. Liu, C. Qian, X. Cao, Post-translational modification control of innate im-
warranted, especially the latter given the rapid increase in micro-
munity, Immunity 45 (2016) 15e30.
bial resistance to antibiotics [87]. Therefore, SUMOs could be a [26] M.M. Hu, Q. Yang, X.Q. Xie, C.Y. Liao, H. Lin, T.T. Liu, et al., Sumoylation pro-
novel target in the fight against infectious and autoimmune motes the stability of the DNA sensor cGAS and the adaptor STING to regulate
the kinetics of response to DNA virus, Immunity 45 (2016) 555e569.
diseases.
[27] M.M. Hu, H.B. Shu, Multifaceted roles of TRIM38 in innate immune and in-
flammatory responses, Cell. Mol. Immunol. 14 (2017) 331e338.
Funding [28] J.H. Kim, K.S. Sung, S.M. Jung, Y.S. Lee, J.Y. Kwon, C.Y. Choi, et al., Pellino-1, an
adaptor protein of interleukin-1 receptor/toll-like receptor signaling, is
sumoylated by Ubc9, Mol. Cells 31 (2011) 85e89.
This work was supported by the Italian “Ministero degli Esteri e [29] M.H. Lee, A.M. Mabb, G.B. Gill, E.T. Yeh, S. Miyamoto, NF-kappaB induction of
della Cooperazione Internazionale” (MAECI); “Associazione Italiana the SUMO protease SENP2: a negative feedback loop to attenuate cell survival
response to genotoxic stress, Mol. Cell 43 (2011) 180e191.
per la ricerca contro il cancro” (AIRC); and Vietnam Ministry of
[30] Y. Zhang, Z. Gan, P. Huang, L. Zhou, T. Mao, M. Shao, et al., A role for protein
Science and Technology (MOST). inhibitor of activated STAT1 (PIAS1) in lipogenic regulation through
SUMOylation-independent suppression of liver X receptors, J. Biol. Chem. 287
(2012) 37973e37985.
Acknowledgements
[31] S.S. Im, T.F. Osborne, Liver x receptors in atherosclerosis and inflammation,
Circ. Res. 108 (2011) 996e1001.
We thank BioScience Writers for editing assistance. [32] W. Huang, S. Ghisletti, K. Saijo, M. Gandhi, M. Aouadi, G.J. Tesz, et al., Coronin
2A mediates actin-dependent de-repression of inflammatory response genes,
Nature 470 (2011) 414e418.
References [33] Y. Ran, T.T. Liu, Q. Zhou, S. Li, A.P. Mao, Y. Li, et al., SENP2 negatively regulates
cellular antiviral response by deSUMOylating IRF3 and conditioning it for
[1] X. Liu, Q. Wang, W. Chen, C. Wang, Dynamic regulation of innate immunity by ubiquitination and degradation, J. Mol. Cell Biol. 3 (2011) 283e292.
ubiquitin and ubiquitin-like proteins, Cytokine & Growth Factor Rev. 24 [34] P. Xia, S. Wang, Z. Xiong, B. Ye, L.Y. Huang, Z.G. Han, et al., IRTKS negatively
(2013) 559e570. regulates antiviral immunity through PCBP2 sumoylation-mediated MAVS
[2] I.A. Hendriks, A.C. Vertegaal, A high-yield double-purification proteomics degradation, Nat. Commun. 6 (2015) 8132.
strategy for the identification of SUMO sites, Nat. Protoc. 11 (2016) [35] T.H. Chang, S. Xu, P. Tailor, T. Kanno, K. Ozato, The small ubiquitin-like
1630e1649. modifier-deconjugating enzyme sentrin-specific peptidase 1 switches IFN
[3] C.M. Hickey, N.R. Wilson, M. Hochstrasser, Function and regulation of SUMO regulatory factor 8 from a repressor to an activator during macrophage acti-
proteases, Nat. Rev. Mol. Cell Biol. 13 (2012) 755e766. vation, J. Immunol. 189 (2012) 3548e3556.
[4] P. Schwertman, S. Bekker-Jensen, N. Mailand, Regulation of DNA double- [36] Q. Liang, H. Deng, X. Li, X. Wu, Q. Tang, T.H. Chang, et al., Tripartite motif-
strand break repair by ubiquitin and ubiquitin-like modifiers, Nat. Rev. Mol. containing protein 28 is a small ubiquitin-related modifier E3 ligase and
28 S. Adorisio et al. / Journal of Autoimmunity 84 (2017) 21e28

negative regulator of IFN regulatory factor 7, J. Immunol. 187 (2011) [63] R.D. Everett, C. Boutell, B.G. Hale, Interplay between viruses and host
4754e4763. sumoylation pathways, Nat. Rev. Microbiol. 11 (2013) 400e411.
[37] T. Kubota, M. Matsuoka, S. Xu, N. Otsuki, M. Takeda, A. Kato, et al., PIASy in- [64] L. Chen, S. Li, Y. Li, X. Duan, B. Liu, I. McGilvray, Ubiquitin-like protein mod-
hibits virus-induced and interferon-stimulated transcription through distinct ifiers and their potential for antiviral and anti-HCV therapy, Expert Rev.
mechanisms, J. Biol. Chem. 286 (2011) 8165e8175. Proteom. 10 (2013) 275e287.
[38] G. Maarifi, M.A. Maroui, J. Dutrieux, L. Dianoux, S. Nisole, M.K. Chelbi-Alix, [65] S. Lefort, A. Gravel, L. Flamand, Repression of interferon-alpha stimulated
Small ubiquitin-like modifier alters IFN response, J. Immunol. 195 (2015) genes expression by Kaposi's sarcoma-associated herpesvirus K-bZIP protein,
2312e2324. Virology 408 (2010) 14e30.
[39] M. Pascual-Garcia, L. Rue, T. Leon, J. Julve, J.M. Carbo, J. Matalonga, et al., [66] J. Gonzalez-Santamaria, M. Campagna, M.A. Garcia, L. Marcos-Villar,
Reciprocal negative cross-talk between liver X receptors (LXRs) and STAT1: D. Gonzalez, P. Gallego, et al., Regulation of vaccinia virus E3 protein by small
effects on IFN-gamma-induced inflammatory responses and LXR-dependent ubiquitin-like modifier proteins, J. Virol. 85 (2011) 12890e12900.
gene expression, J. Immunol. 190 (2013) 6520e6532. [67] S. Liu, J. Long, B. Yuan, M. Zheng, M. Xiao, J. Xu, et al., SUMO modification
[40] K. Doiron, V. Goyon, E. Coyaud, S. Rajapakse, B. Raught, H.M. McBride, The reverses inhibitory effects of smad nuclear interacting Protein-1 in TGF-beta
dynamic interacting landscape of MAPL reveals essential functions for responses, J. Biol. Chem. 291 (2016) 24418e24430.
SUMOylation in innate immunity, Sci. Rep. 7 (2017) 107. [68] P.C. Chang, M. Campbell, E.S. Robertson, Human oncogenic herpesvirus and
[41] X. Ding, S. Jin, Y. Tong, X. Jiang, Z. Chen, S. Mei, et al., TLR4 signaling induces post-translational modifications - phosphorylation and SUMOylation, Front.
TLR3 up-regulation in alveolar macrophages during acute lung injury, Sci. Rep. Microbiol. 7 (2016) 962.
7 (2017) 34278. [69] J. Gan, N. Qiao, R. Strahan, C. Zhu, L. Liu, S.C. Verma, et al., Manipulation of
[42] J. Zhang, Z. Chen, Z. Zhou, P. Yang, C.Y. Wang, Sumoylation modulates the ubiquitin/SUMO pathways in human herpesviruses infection, Rev. Med. Virol.
susceptibility to type 1 diabetes, Adv. Exp. Med. Biol. 963 (2017) 299e322. 26 (2016) 435e445.
[43] C. Sun, H. Wei, X. Chen, Z. Zhao, H. Du, W. Song, et al., ERBB3-rs2292239 as [70] G.L. Bentz, J. Shackelford, J.S. Pagano, Epstein-Barr virus latent membrane
primary type 1 diabetes association locus among non-HLA genes in Chinese, protein 1 regulates the function of interferon regulatory factor 7 by inducing
Meta Gene 9 (2016) 120e123. its sumoylation, J. Virol. 86 (2012) 12251e12261.
[44] K.W. Hwang, T.J. Won, H. Kim, H.J. Chun, T. Chun, Y. Park, Characterization of [71] C. Fraisier, R. Rodrigues, V. Vu Hai, M. Belghazi, S. Bourdon, G. Paranhos-
the regulatory roles of the SUMO, Diabetes/Metab. Res. Rev. 27 (2011) Baccala, et al., Hepatocyte pathway alterations in response to in vitro Crimean
854e861. Congo hemorrhagic fever virus infection, Virus Res. 179 (2014) 187e203.
[45] K.W. Hwang, T.J. Won, H. Kim, H.J. Chun, T. Chun, Y. Park, Erratum to 'Char- [72] L. Marcos-Villar, J.V. Perez-Giron, J.M. Vilas, A. Soto, C.F. de la Cruz-Hererra,
acterization of the regulatory roles of the SUMO, Diabetes/Metab. Res. Rev. 28 V. Lang, et al., SUMOylation of p53 mediates interferon activities, Cell Cycle 12
(2012) 196e202. (2013) 2809e2816.
[46] L. Shao, B. Feng, Y. Zhang, H. Zhou, W. Ji, W. Min, The role of adipose-derived [73] G. Maarifi, Z. Hannoun, M.C. Geoffroy, F. El Asmi, K. Zarrouk, S. Nisole, et al.,
inflammatory cytokines in type 1 diabetes, Adipocyte 5 (2016) 270e274. MxA mediates SUMO-induced resistance to vesicular stomatitis virus, J. Virol.
[47] D. Sireesh, E. Bhakkiyalakshmi, K.M. Ramkumar, S. Rathinakumar, 90 (2016) 6598e6610.
P.S. Jennifer, P. Rajaguru, et al., Targeting SUMOylation cascade for diabetes [74] P.C. Chang, C.Y. Cheng, M. Campbell, Y.C. Yang, H.W. Hsu, T.Y. Chang, et al., The
management, Curr. Drug Targets 15 (2014) 1094e1106. chromatin modification by SUMO-2/3 but not SUMO-1 prevents the epige-
[48] G. Park, H.S. Kim, J.Y. Choe, S.K. Kim, SUMO4 C438T polymorphism is asso- netic activation of key immune-related genes during Kaposi's sarcoma asso-
ciated with papulopustular skin lesion in Korean patients with Behcet's dis- ciated herpesvirus reactivation, BMC Genomics 14 (2013) 824.
ease, Rheumatol. Int. 32 (2012) 3031e3037. [75] E. Sloan, M.H. Tatham, M. Groslambert, M. Glass, A. Orr, R.T. Hay, et al.,
[49] F. Li, X. Li, L. Kou, Y. Li, F. Meng, F. Ma, SUMO-conjugating enzyme UBC9 Analysis of the SUMO2 proteome during HSV-1 infection, PLoS Pathog. 11
promotes proliferation and migration of fibroblast-like synoviocytes in (2015), e1005059.
rheumatoid arthritis, Inflammation 37 (2014) 1134e1141. [76] S. Callegari, S. Gastaldello, O.R. Faridani, M.G. Masucci, Epstein-Barr virus
[50] K.D. Preuss, M. Pfreundschuh, M. Weigert, N. Fadle, E. Regitz, B. Kubuschok, encoded microRNAs target SUMO-regulated cellular functions, FEBS J. 281
Sumoylated HSP90 is a dominantly inherited plasma cell dyscrasias risk fac- (2014) 4935e4950.
tor, J. Clin. Investig. 125 (2015) 316e323. [77] N. Reuter, E.M. Schilling, M. Scherer, R. Muller, T. Stamminger, The ND10
[51] K. Sarkar, S. Sadhukhan, S.S. Han, Y.M. Vyas, SUMOylation-disrupting WAS component promyelocytic leukemia protein acts as an E3 ligase for SUMOy-
mutation converts WASp from a transcriptional activator to a repressor of NF- lation of the major immediate early protein IE1 of human cytomegalovirus,
kappaB response genes in T cells, Blood 126 (2015) 1670e1682. J. Virol. (2017) 91.
[52] M.C. Maroun, O. Olivero, L. Lipovich, A. Stark, L. Tait, S. Bandyopadhyay, et al., [78] J. Berscheminski, P. Wimmer, J. Brun, W.H. Ip, P. Groitl, T. Horlacher, et al.,
Anti-centrosome antibodies in breast cancer are the expression of autoim- Sp100 isoform-specific regulation of human adenovirus 5 gene expression,
munity, Immunol. Res. 60 (2014) 339e347. J. Virol. 88 (2014) 6076e6092.
[53] M. Gou, Q. Huang, W. Qian, Z. Zhang, Z. Jia, J. Hua, Sumoylation E3 ligase SIZ1 [79] Y. Izumiya, K. Kobayashi, K.Y. Kim, M. Pochampalli, C. Izumiya, B. Shevchenko,
modulates plant immunity partly through the immune receptor gene SNC1 in et al., Kaposi's sarcoma-associated herpesvirus K-Rta exhibits SUMO-targeting
Arabidopsis, Mol. Plant-Microbe Interact. MPMI 30 (2017) 334e342. ubiquitin ligase (STUbL) like activity and is essential for viral reactivation,
[54] D. Ribet, M. Hamon, E. Gouin, M.A. Nahori, F. Impens, H. Neyret-Kahn, et al., PLoS Pathog. 9 (2013), e1003506.
Listeria monocytogenes impairs SUMOylation for efficient infection, Nature [80] P.C. Chang, H.J. Kung, SUMO and KSHV replication, Cancers 6 (2014)
464 (2010) 1192e1195. 1905e1924.
[55] H. Pillich, T. Chakraborty, M.A. Mraheil, Cell-autonomous responses in Listeria [81] T. Gunther, S. Schreiner, T. Dobner, U. Tessmer, A. Grundhoff, Influence of
monocytogenes infection, Future Microbiol. 10 (2015) 583e597. ND10 components on epigenetic determinants of early KSHV latency estab-
[56] K. Jo, E.J. Kim, H.J. Yu, C.H. Yun, D.W. Kim, Host cell nuclear localization of lishment, PLoS Pathog. 10 (2014), e1004274.
Shigella flexneri effector OspF is facilitated by SUMOylation, J. Microbiol. [82] Z. Mi, J. Fu, Y. Xiong, H. Tang, SUMOylation of RIG-I positively regulates the
Biotechnol. 27 (2017) 610e615. type I interferon signaling, Protein & Cell 1 (2010) 275e283.
[57] S.M. Sidik, J. Salsman, G. Dellaire, J.R. Rohde, Shigella infection interferes with [83] D. Lopez-Farfan, J.M. Bart, D.I. Rojas-Barros, M. Navarro, SUMOylation by the
SUMOylation and increases PML-NB number, PLoS One 10 (2015), e0122585. E3 ligase TbSIZ1/PIAS1 positively regulates VSG expression in Trypanosoma
[58] J.G. Kim, W. Stork, M.B. Mudgett, Xanthomonas type III effector XopD desu- brucei, PLoS Pathog. 10 (2014), e1004545.
moylates tomato transcription factor SlERF4 to suppress ethylene responses [84] N. Pozzesi, A. Fierabracci, T.T. Thuy, M.P. Martelli, A.M. Liberati, E. Ayroldi, et
and promote pathogen growth, Cell Host Microbe 13 (2013) 143e154. al., Pharmacological modulation of caspase-8 in thymus-related medical
[59] C.M. Tan, M.Y. Li, P.Y. Yang, S.H. Chang, Y.P. Ho, H. Lin, et al., Arabidopsis HFR1 conditions, J. Pharmacol. Exp. Ther. 351 (2014) 18e24.
is a potential nuclear substrate regulated by the Xanthomonas type III effector [85] D.V. Delfino, S. Spinicelli, N. Pozzesi, S. Pierangeli, E. Velardi, S. Bruscoli, et al.,
XopD(Xcc8004), PLoS One 10 (2015), e0117067. Glucocorticoid-induced activation of caspase-8 protects the glucocorticoid-
[60] S.B. Smith, M. Magid-Slav, J.R. Brown, Host response to respiratory bacterial induced protein Gilz from proteasomal degradation and induces its binding
pathogens as identified by integrated analysis of human gene expression data, to SUMO-1 in murine thymocytes, Cell Death Differ. 18 (2011) 183e190.
PLoS One 8 (2013), e75607. [86] N. Pozzesi, A. Fierabracci, A.M. Liberati, M.P. Martelli, E. Ayroldi, C. Riccardi, et
[61] D. Mattoscio, C.V. Segre, S. Chiocca, Viral manipulation of cellular protein al., Role of caspase-8 in thymus function, Cell Death Differ. 21 (2014)
conjugation pathways: the SUMO lesson, World J. Virol. 2 (2013) 79e90. 226e233.
[62] V.G. Wilson, Sumoylation at the host-pathogen interface, Biomolecules 2 [87] D.R. Dodds, Antibiotic resistance: a current epilogue, Biochem. Pharmacol. 134
(2012) 203e227. (2017) 139e146.

Vous aimerez peut-être aussi