Inmunidad Innata

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Available online at www.sciencedirect.

com

ScienceDirect

Innate immunity signalling and membrane trafficking


Tomohiko Taguchi1,2 and Kojiro Mukai1

The mammalian innate immune system serves as the front line receptors [4], nucleotide-binding domain and leucine-
of the host to eliminate invading pathogens. The receptors that rich repeat-containing receptors (NLRs) [5], C-type lec-
sense invading pathogens or the pathogen-associated tin receptors (CLRs) [6], cyclic GMP-AMP (cGAMP)
molecules localized at various membrane compartments that synthase [7], and stimulator of interferon genes (STING)
include the plasma membrane, endosomes, and the [8].
endoplasmic reticulum. Intriguingly, growing evidence
indicates that the sites of pathogen detection do not always Several PRRs, such as TLRs, CLRs, and STING, are
represent the site where innate immune signal is triggered. transmembrane proteins and distribute at various mem-
Rather, pathogen detection often induces translocation of the brane compartments, such as the plasma membrane
receptors by membrane trafficking. Furthermore, dysregulated (PM), early endosomes, late endosomes, recycling endo-
membrane trafficking of the receptors renders the host somes (REs), and the endoplasmic reticulum (ER). Some
susceptible to infection or prone to autoinflammatory diseases. PRRs, upon binding to PAMPs, translocate to other
These findings underscore the critical role of membrane compartments by membrane trafficking and triggers
trafficking in the innate immunity. In this review, we highlight the innate immunity signalling there. Not surprisingly,
emerging issues regarding PRRs and membrane trafficking, dysregulated membrane trafficking of PRRs often under-
with the particular focus on STING and TLR4, the activity of lies the pathogenesis of autoinflammatory diseases. This
which is tightly regulated by membrane trafficking. article is not intended as a comprehensive review of the
innate immunity signalling and PRRs. Rather, we aim to
Addresses
1
highlight recent publications and emerging issues regard-
Laboratory of Organelle Pathophysiology, Department of Integrative ing PRRs and membrane trafficking, with the particular
Life Sciences, Graduate School of Life Sciences, Tohoku University,
Aobayama, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
focus on STING and TLR4, the activity of which is
2
AMED-PRIME, Japan Agency for Medical Research and Development, revealed to be tightly coupled with membrane trafficking.
1-7-1, Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan
Exocytic trafficking of STING from the ER
Corresponding author: Taguchi, Tomohiko (tomohiko.taguchi.
b8@tohoku.ac.jp)
DNA is normally present in the nucleus and mitochondria
of eukaryotic cells, and the presence of DNA in aberrant
locations, such as cytosol, is believed to trigger immune
Current Opinion in Cell Biology 2019, 59:1–7 response [9]. Indeed, recent studies have identified two
This review comes from a themed issue on Membrane Trafficking central PRRs, cGAMP synthase (cGAS) [7] and STING
Edited by Akihiko Nakano and Peter Cullen [8], which are required for the immune response against
cytosolic DNA. DNA viruses and bacteria can release
their genomic dsDNA to the host cytoplasm during
infection. The cytosolic dsDNA activates cGAS, leading
https://doi.org/10.1016/j.ceb.2019.02.002 to the generation of cGAMP, a cyclic dinucleotide
0955-0674/ã 2018 Elsevier Ltd. All rights reserved. (CDN), from GTP and ATP [10]. cGAMP then activates
STING to trigger the type I interferon responses [11].
Other CDNs including cyclic di-GMP [12] and cyclic di-
AMP [13] are secreted during intracellular bacterial infec-
tion, and they directly activate STING [11,14].

Introduction STING [8], also known as MITA [15], ERIS [16], MPYS
Vertebrates have evolved biological systems to combat [17], or TMEM173, is an ER-localized transmembrane
invading pathogens. As the first line of host defense, the protein (Figure 1). Mechanistically, STING serves as a
innate immune system recognizes microbial pathogens scaffold for TBK1 kinase and the transcription factor
with host germline-encoded pattern recognition receptors interferon regulatory factor 3 (IRF3) [18,19]. IRF3 is
(PRRs) that bind unique pathogen-associated molecular phosphorylated by TBK1, after which it translocates into
patterns (PAMPs) [1,2]. Activated PRRs triggers intracel- the nucleus to stimulate the transcription of type I inter-
lular signalling cascades, which lead to transcriptional ferons such as interferon b [20]. Interestingly, after the
expression of proinflammatory cytokines, type I interfer- binding of STING to CDNs, STING immediately trans-
ons, and other antiviral proteins that all coordinate the locates from the ER to perinuclear compartments that
elimination of pathogens and infected cells. PRRs include the Golgi, endosomes, and autophagy-related
include Toll-like receptors (TLRs) [3], RIG-I-like compartments [21,22]. The exocytic membrane

www.sciencedirect.com Current Opinion in Cell Biology 2019, 59:1–7

Descargado para Anonymous User (n/a) en Universidad Simon Bolivar de ClinicalKey.es por Elsevier en abril 19, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
2 Membrane Trafficking

Figure 1

DNA virus Cell membrane

ATP Genomic DNA


+ cGAS
GTP Mitochondrial DNA
cGAMP
(cyclic GMP-AMP)
P P
Bacteria IRF3 TBK1
STING STING STING STING
CDNs Endoplasmic
(cyclic dinucleotides) reticulum
Palmitoylation
Nucleus
Cys88/91

P IFNβ Golgi Recycling endosomes Lysosomes


IRF3

Antiviral response
Autoinflammatory diseases

Current Opinion in Cell Biology

Regulation of the cGAS-STING signalling pathway by membrane trafficking.


The cytosolic dsDNA derived from DNA virus, host genome, or mitochondria activates cGAS, leading to the generation of cGAMP. cGAMP directly
binds to STING and induces translocation of STING from the ER to the Golgi. STING undergoes palmitoylation at the Golgi which is essential for
the activation of STING. After exit from the Golgi, STING further moves to REs and then to the p62-positive compartments/lysosomes.

trafficking of STING may be essential for the STING- c-di-GMP binding [30,31] or cGAMP binding [32,33].
dependent signalling events, because treatment with In brief, cGAMP binding induced the STING dimer to
brefeldin A (BFA) or expression of Shigella effector form a closed conformation, resulting in a deeper cGAMP
IpaJ, which blocks ER-to-Golgi traffic, abolished binding pocket that is covered with a b sheet lid.
phosphorylation of IRF3 and induction of interferon b
[21,23,24,25]. More direct evidence was provided by the These structural analyses provided a detailed picture of
recent observation that knockdown of Sar1, a small how STING recognizes CDNs. However, it is still not
GTPase that regulates coat protein complex II (COP- clear how the conformational change associated with the
II)-mediated ER-to-Golgi traffic, suppressed phosphory- CDN binding leads to the translocation of STING from
lation of IRF3 [26]. the ER to the Golgi with the COP-II complex, and
eventually to the activation of the STING-TBK1-IRF3
Structural insight of STING activation signalling pathway. The very recent study has even
Several crystal structures of the STING cytoplasmic casted doubt on the idea that the closed conformation
domain and its complex with CDNs were elucidated. of STING dimer is critical for activation [34]. In the
Three independent crystal structures of STING bound to study, a linked amidobenzimidazoles-based compound
cyclic di-GMP did not reveal any major structural changes (diABZI-compound 2) was developed. This novel non-
following c-di-GMP binding [27–29]. However, these nucleotide compound efficiently activated STING with
studies used a rare human STING variant, R232H, which maintaining an open STING dimer conformation. So far,
is partially defective in stimulating type I interferons. only the conformation of the C-terminal cytoplasmic
Other structural studies, which used wild-type STING domain of STING was analyzed. Structural elucidation
(R232), revealed a conformational change induced by of full-length STING, which includes N-terminal

Current Opinion in Cell Biology 2019, 59:1–7 www.sciencedirect.com

Descargado para Anonymous User (n/a) en Universidad Simon Bolivar de ClinicalKey.es por Elsevier en abril 19, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Innate immunity signalling and membrane trafficking Taguchi and Mukai 3

Figure 2

Transmembrane domain Cyclic dinucleotide-Binding Domain C-Terminal Tail


1 21 41 47 67 87 106 116 136 151 341 379

C88/C91 human: V147L/M human: N154S human: V155M human: C206Y human: R281Q human: R284G/S
(Palmitoylation) (mouse: V146L/M) (mouse: N153S) (mouse: V154M) (mouse: C205Y) (mouse: R280Q) (mouse: R283G/S)
Y. Liu et al., N Engl J Med 371, 507 (2014). I. Melki et al., J Allergy Clin Immunol 140, 543 (2017).
J. Munoz et al., JAMA Dermatol 151, 872 (2015). H. Konno et al., Cell Rep 23, 1112 (2018).

cGAMP

90°

cytosol

lumen
Current Opinion in Cell Biology

Schematic overview of STING and localization of mutations that cause SAVI.


V147/N154/V155 (shown in red), but not C206/R281/R284 (shown in blue), lie in the domain that is involved in STING dimerization.

transmembrane domains, will help understand the molec- variants required the exocytic membrane traffic from
ular mechanism of STING activation with CDNs. the ER for their activity. In contrast to wild-type
STING, how SAVI variants can get access to the exo-
STING-associated disease cytic membrane traffic without cGAMP stimulation is
Gain-of-function mutations in STING have been asso- not understood.
ciated with an autoinflammatory syndrome termed
STING-associated vasculopathy with onset in infancy Interestingly, 4 variants (C206Y, R281Q, R284G, and
(SAVI) (Figure 2) [35–38]. SAVI is characterized by R284S) [38,39] that were recently identified did not lie
early-onset systemic inflammation with fever, a severe in the domain that is involved in STING dimerization. A
skin vasculopathy that leads to extensive tissue loss in recent survey of STING mutations in the Catalogue of
some cases, and interstitial lung disease resulting in Somatic Mutations in Cancer database led to the demon-
pulmonary fibrosis and end-stage respiratory failure. stration that R284M mutation was able to hyperactivate
The SAVI mutations (V147L, N154S, V155M, C206Y, STING [40]. Alternative mutation of R284K or R284T
R281Q, and R284G) are shown to be associated with also hyperactivates STING. Thus, multiple types of
constitutive activation of STING without DNA and amino acid substitution at 284 confer a gain-of-function
cGAMP stimulation. These SAVI variants did not local- mutation, indicating a high degree of specificity for the
ize at the ER, instead localized at the perinuclear com- function of R284. Given that R284 is exposed at the
partments [26]. The suppression of ER-to-Golgi traffic surface of STING, R284 may be involved in protein
abolished TBK1 binding to the SAVI variants [26], interactions by which the ER localization of STING is
suggesting that even the constitutively active SAVI maintained.

www.sciencedirect.com Current Opinion in Cell Biology 2019, 59:1–7

Descargado para Anonymous User (n/a) en Universidad Simon Bolivar de ClinicalKey.es por Elsevier en abril 19, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
4 Membrane Trafficking

STING activation at the Golgi binds downstream adaptor proteins. Much of our current
As described above, the post-ER membrane compart- knowledge of PAMP-induced PRR trafficking has been
ments are required for activation of STING. Recent derived from the study of TLR4. TLR4 binds extracel-
evidence suggested that palmitoylation of STING at lular lipopolysaccharide (LPS), the major outer mem-
the Golgi is essential for activation of STING [23]. brane component of Gram-negative bacteria. After
Treatment with protein palmitoylation inhibitor 2-bro- binding to LPS at the PM, TLR4 interacts with the
mopalmitate (2-BP) abolished STING-dependent type I PtdIns(4,5)P2-effector TIRAP and the associated
interferon response. Mutation of two membrane-proximal MyD88, which results in induction of the secretion of
Cys residues (C88/91) suppressed palmitoylation, and this proinflammatory cytokines [48]. After translocation to
STING mutant could not induce STING-dependent early endosomes/phagosomes, the complex disassembles
host defense genes. The response elicited by the SAVI most likely due to the low concentration of PtdIns(4,5)
variants was also effectively inhibited by 2-BP or an P2. This allows other adaptors TRAM and TRIF to bind
introduction of C88/91S mutation. The importance of to TLR4, which triggers secretion of the type I interfer-
C88/91 in STING activation was further emphasized ons [49].
by the recent findings of identification of STING inhi-
bitors that target C88/91 of STING [41,42]. A role of Rab11-positive REs in TLR4 signalling has
been demonstrated [50,51]. Endogenous TLR4 localizes
Protein palmitoylation has been implicated in the clus- mostly in Rab11-positive REs in unstimulated human
tering of a number of proteins [43] such as H-ras and Fas monocytes. By stimulation with Escherichia coli, a Gram-
into lipid rafts (specific membrane domains enriched in negative bacterium, TLR4 was transported from REs to
cholesterol and sphingomyelin (SM)). Cholesterol is sug- E. coli-positive phagosomes. Knockdown of Rab11
gested to be enriched at the TGN, and cholesterol resulted in emptying of TLR4 in REs, reduction of
together with SM generated by SM synthase 1 are TLR4 around phagosomes, and inhibition of E. coli-
thought to form lipid rafts at the TGN [44]. Treatment induced IRF3 activation. Signalling lymphocytic activa-
of cells with C6-ceramide disrupted lipid rafts at the Golgi tion molecule family 1 (SLAMF1) is an Ig-like receptor
by generating short-chain SM that disturbs the lipid order and a costimulatory molecule that initiates signal trans-
[44]. C6-ceramide inhibited the STING activity without duction networks in a variety of immune cells. In resting
affecting the translocation of STING to the Golgi [23]. macrophages, SLAMF1 was localized to REs, but upon
Therefore, STING palmitoylation may allow clustering addition of E. coli, it was trafficked together with TRAM
of STING at the lipid rafts of the TGN, which then from REs to E. coli-positive phagosomes in a Rab11-
facilitates the recruitment and interaction of downstream dependent manner. Thus, the membrane traffic from
signalling factors such as TBK1 and IRF3 on STING. REs helps fuel phagosomes with TLR4 and its effector
protein for maximal TLR4 signalling (Figure 3). Rab11
Post-Golgi trafficking of STING also functioned in maintaining proper subcellular locali-
After exit from the Golgi, STING moved to REs and then zation of endosomal TLR9 [52].
to the p62-positive compartments/lysosomes [22,23,45].
STING appears to be degraded eventually at lysosomes TLR4 degradation at lysosomes
because chloroquine or bafilomycin A1, which interferes VPS33B is the homolog of yeast class C vacuolar protein
with lysosomal functions, prevented STING degradation sorting (vps) protein Vps33p. Class C vps proteins func-
[24,46]. Bafilomycin A1 treatment enhanced STING- tion in at least two multiprotein complexes, class C core
dependent host defense genes [46], suggesting that deg- vacuole/endosome tethering (CORVET) and homotypic
radation of STING at lysosomes is required to terminate fusion and vacuole protein sorting (HOPS), which inter-
the STING signalling. The palmitoylation of STING at act with yeast orthologs of Rab5 and Rab7 GTPases and
the Golgi was not required for the degradation process, are crucial for several steps in vesicular trafficking path-
because the palmitoylation-deficient STING mutant ways leading to lysosomes. In a recent study, Drosophila
(C88/91S) underwent degradation like wild-type STING and mammalian Vps33B proteins were shown to play
[23]. The exact molecular mechanism underlying critical roles in the degradation of TLR4 after LPS
STING trafficking from endosomes to lysosomes is not stimulation [53]. TLR4 accumulated at Rab5-positive,
clear. There have been controversies regarding the con- Rab7-positive, or Rab11-positive endosomes in Vps33B-
tribution of the autophagic process to degradation of silenced macrophages. The prolonged residency of TLR4
STING [45–47]. in these endosomes paralleled well with the enhanced
expression of proinflammatory cytokines. Lack of Vps33B
Membrane trafficking of TLR4 had no effect on trafficking of non-microbial cargo.
TLRs are type I transmembrane proteins that are com-
posed of three distinct structural domains: a leucine-rich Arthrogryposis, renal dysfunction, and cholestasis syn-
repeat containing ectodomain involved in PAMP binding, drome (ARC) is a multisystem disorder associated with
a transmembrane domain, and a cytosolic domain that abnormalities in polarized liver and kidney cells.

Current Opinion in Cell Biology 2019, 59:1–7 www.sciencedirect.com

Descargado para Anonymous User (n/a) en Universidad Simon Bolivar de ClinicalKey.es por Elsevier en abril 19, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Innate immunity signalling and membrane trafficking Taguchi and Mukai 5

Figure 3

Gram-negative bacteria

LPS TLR4

PI(4,5)P2
TIRAP Phagocytosis
MyD88
Endocytosis

Rab11

TRAM Early TRAM Phagosomes Recycling


TRIF endosomes TRIF endosomes

Induction of type I interferons

Induction of proinflammatory cytokines

Current Opinion in Cell Biology

Regulation of the TLR4 signalling pathway by membrane trafficking.

Persistent infections and sepsis are recurring symptoms in PRRs trigger distinct signalling pathways, and the molec-
ARC patients. Germline mutations in VPS33B are found ular mechanism of how PRR signals are terminated.
in approximately 75% of individuals with ARC [54]. These studies are anticipated to make broad conceptual
Exaggerated inflammatory responses induced by persis- contributions, beyond the innate immunity field, to the
tence of PRRs in aberrant endosomal compartments may understanding of essentials of membrane traffic and cell
contribute to symptoms of ARC syndrome. biology.

Concluding remarks
The last few decades have witnessed substantial progress Conflict of interest statement
in the field of innate immunity since the identification of Nothing declared.
the first mammalian TLRs in the late 1990s. Technical
advances in cell biology, biochemistry, and molecular
biology have facilitated our understanding of the regula- Author contribution statement
tion of PRR signalling. The importance of understanding Draft and funding acquisition: T.T. and K.M.; Concep-
cell biological aspects of innate immunity has been tualization and writing: T.T.
emphasized by the fact that some autoinflammatory
and autoimmune diseases are caused by mislocalization
of PRRs. Acknowledgements
K.M. is supported by Japan Society for the Promotion of Science (JSPS)
Despite these advances, many questions remain unan- KAKENHI Grant JP17K15445, Mochida Memorial Foundation for Medical
and Pharmaceutical Research, and MSD Life Science Foundation, Public
swered regarding the regulators of membrane trafficking Interest Incorporated Foundation. T.T. is supported by JSPS KAKENHI
of PRRs, the nature of various endosomes from which Grants JP16H04782, JP15H05903, and AMED-PRIME.

www.sciencedirect.com Current Opinion in Cell Biology 2019, 59:1–7

Descargado para Anonymous User (n/a) en Universidad Simon Bolivar de ClinicalKey.es por Elsevier en abril 19, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
6 Membrane Trafficking

References and recommended reading 19. Tanaka Y, Chen ZJ: STING specifies IRF3 phosphorylation by
TBK1 in the cytosolic DNA signaling pathway. Sci Signal 2012,
Papers of particular interest, published within the period of review, 5:ra20.
have been highlighted as:
20. Fitzgerald KA, McWhirter SM, Faia KL, Rowe DC, Latz E,
 of special interest Golenbock DT, Coyle AJ, Liao SM, Maniatis T: IKKepsilon and
 of outstanding interest TBK1 are essential components of the IRF3 signaling pathway.
Nat Immunol 2003, 4:491-496.
1. Palm NW, Medzhitov R: Pattern recognition receptors and
21. Ishikawa H, Ma Z, Barber GN: STING regulates intracellular
control of adaptive immunity. Immunol Rev 2009, 227:221-233.
DNA-mediated, type I interferon-dependent innate immunity.
2. Takeuchi O, Akira S: Pattern recognition receptors and Nature 2009, 461:788-792.
inflammation. Cell 2010, 140:805-820. 22. Saitoh T, Fujita N, Hayashi T, Takahara K, Satoh T, Lee H,
Matsunaga K, Kageyama S, Omori H, Noda T et al.: Atg9a
3. Blasius AL, Beutler B: Intracellular toll-like receptors. Immunity controls dsDNA-driven dynamic translocation of STING and
2010, 32:305-315. the innate immune response. Proc Natl Acad Sci U S A 2009,
106:20842-20846.
4. Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T,
Miyagishi M, Taira K, Akira S, Fujita T: The RNA helicase RIG-I 23. Mukai K, Konno H, Akiba T, Uemura T, Waguri S, Kobayashi T,
has an essential function in double-stranded RNA-induced  Barber GN, Arai H, Taguchi T: Activation of STING requires
innate antiviral responses. Nat Immunol 2004, 5:730-737. palmitoylation at the Golgi. Nat Commun 2016, 7:11932.
This study shows that palmitoylation of STING at the Golgi is essential for
5. Ye Z, Ting JP: NLR, the nucleotide-binding domain leucine-rich
the activation of STING, thus answering the essential question of why
repeat containing gene family. Curr Opin Immunol 2008, 20:3-9.
STING needs translocation from the ER for its activation by membrane
6. Geijtenbeek TB, Gringhuis SI: Signalling through C-type lectin trafficking. The authors show that the activity of SAVI variants is effec-
receptors: shaping immune responses. Nat Rev Immunol 2009, tively inhibited by 2-bromopalmitate or an introduction of Cys88/91Ser
9:465-479. mutation. These results may lead to new treatments for cytosolic DNA-
triggered autoinflammatory diseases.
7. Sun L, Wu J, Du F, Chen X, Chen ZJ: Cyclic GMP-AMP synthase
is a cytosolic DNA sensor that activates the type I interferon 24. Konno H, Konno K, Barber GN: Cyclic dinucleotides trigger
pathway. Science 2013, 339:786-791. ULK1 (ATG1) phosphorylation of STING to prevent sustained
innate immune signaling. Cell 2013, 155:688-698.
8. Ishikawa H, Barber GN: STING is an endoplasmic reticulum
25. Dobbs N, Burnaevskiy N, Chen D, Gonugunta VK, Alto NM, Yan N:
adaptor that facilitates innate immune signalling. Nature 2008,
STING activation by translocation from the ER is associated
455:674-678.
with infection and autoinflammatory disease. Cell Host Microbe
9. Stetson DB, Medzhitov R: Recognition of cytosolic DNA 2015, 18:157-168.
activates an IRF3-dependent innate immune response. 26. Ogawa E, Mukai K, Saito K, Arai H, Taguchi T: The binding of
Immunity 2006, 24:93-103.  TBK1 to STING requires exocytic membrane traffic from the
10. Wu J, Sun L, Chen X, Du F, Shi H, Chen C, Chen ZJ: Cyclic GMP- ER. Biochem Biophys Res Commun 2018, 503:138-145.
AMP is an endogenous second messenger in innate immune This study shows that the binding of TBK1 to STING, a critical step before
signaling by cytosolic DNA. Science 2013, 339:826-830. phosphorylation of TBK1, occurs at the Golgi. Curiously, even constitu-
tively active SAVI variants cannot bind to TBK1 when these STING
11. Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B, variants are trapped in the ER. The authors suggest that the Golgi is
Hyodo M, Hayakawa Y, Vance RE: STING is a direct innate an organelle at which STING recruits and activates TBK1.
immune sensor of cyclic di-GMP. Nature 2011, 478:515-518.
27. Yin Q, Tian Y, Kabaleeswaran V, Jiang X, Tu D, Eck MJ, Chen ZJ,
12. McWhirter SM, Barbalat R, Monroe KM, Fontana MF, Hyodo M, Wu H: Cyclic di-GMP sensing via the innate immune signaling
Joncker NT, Ishii KJ, Akira S, Colonna M, Chen ZJ et al.: A host protein STING. Mol Cell 2012, 46:735-745.
type I interferon response is induced by cytosolic sensing of 28. Shu C, Yi G, Watts T, Kao CC, Li P: Structure of STING bound to
the bacterial second messenger cyclic-di-GMP. J Exp Med cyclic di-GMP reveals the mechanism of cyclic dinucleotide
2009, 206:1899-1911. recognition by the immune system. Nat Struct Mol Biol 2012,
13. Woodward JJ, Iavarone AT, Portnoy DA: c-di-AMP secreted by 19:722-724.
intracellular Listeria monocytogenes activates a host type I 29. Ouyang S, Song X, Wang Y, Ru H, Shaw N, Jiang Y, Niu F, Zhu Y,
interferon response. Science 2010, 328:1703-1705. Qiu W, Parvatiyar K et al.: Structural analysis of the STING
adaptor protein reveals a hydrophobic dimer interface and
14. Barker JR, Koestler BJ, Carpenter VK, Burdette DL, Waters CM,
mode of cyclic di-GMP binding. Immunity 2012, 36:1073-1086.
Vance RE, Valdivia RH: STING-dependent recognition of cyclic
di-AMP mediates type I interferon responses during 30. Shang G, Zhu D, Li N, Zhang J, Zhu C, Lu D, Liu C, Yu Q, Zhao Y,
Chlamydia trachomatis infection. mBio 2013, 4:e00018-13. Xu S, Gu L: Crystal structures of STING protein reveal basis for
recognition of cyclic di-GMP. Nat Struct Mol Biol 2012, 19:725-
15. Zhong B, Yang Y, Li S, Wang YY, Li Y, Diao F, Lei C, He X, Zhang L,
727.
Tien P, Shu HB: The adaptor protein MITA links virus-sensing
receptors to IRF3 transcription factor activation. Immunity 31. Huang YH, Liu XY, Du XX, Jiang ZF, Su XD: The structural basis
2008, 29:538-550. for the sensing and binding of cyclic di-GMP by STING. Nat
Struct Mol Biol 2012, 19:728-730.
16. Sun W, Li Y, Chen L, Chen H, You F, Zhou X, Zhou Y, Zhai Z,
Chen D, Jiang Z: ERIS, an endoplasmic reticulum IFN 32. Zhang X, Shi H, Wu J, Zhang X, Sun L, Chen C, Chen ZJ: Cyclic
stimulator, activates innate immune signaling through GMP-AMP containing mixed phosphodiester linkages is an
dimerization. Proc Natl Acad Sci U S A 2009, 106:8653-8658. endogenous high-affinity ligand for STING. Mol Cell 2013,
51:226-235.
17. Jin L, Waterman PM, Jonscher KR, Short CM, Reisdorph NA,
Cambier JC: MPYS, a novel membrane tetraspanner, is 33. Gao P, Ascano M, Zillinger T, Wang W, Dai P, Serganov AA,
associated with major histocompatibility complex class II and Gaffney BL, Shuman S, Jones RA, Deng L et al.: Structure-
mediates transduction of apoptotic signals. Mol Cell Biol 2008, function analysis of STING activation by c[G(20 ,50 )pA(30 ,50 )p]
28:5014-5026. and targeting by antiviral DMXAA. Cell 2013, 154:748-762.
18. Liu S, Cai X, Wu J, Cong Q, Chen X, Li T, Du F, Ren J, Wu YT, 34. Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R,
Grishin NV, Chen ZJ: Phosphorylation of innate immune  Zhang SY, Tran JL, Moore P, Lehmann S, Eberl HC et al.: Design
adaptor proteins MAVS, STING, and TRIF induces IRF3 of amidobenzimidazole STING receptor agonists with
activation. Science 2015, 347:aaa2630. systemic activity. Nature 2018, 564:439-443.

Current Opinion in Cell Biology 2019, 59:1–7 www.sciencedirect.com

Descargado para Anonymous User (n/a) en Universidad Simon Bolivar de ClinicalKey.es por Elsevier en abril 19, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Innate immunity signalling and membrane trafficking Taguchi and Mukai 7

This study identifies a small molecule human STING agonist that is not a 44. Duran JM, Campelo F, van Galen J, Sachsenheimer T, Sot J,
CDN. This novel non-nucleotide compound efficiently activates STING Egorov MV, Rentero C, Enrich C, Polishchuk RS, Goñi FM et al.:
with maintaining an open STING dimer conformation, suggesting a Sphingomyelin organization is required for vesicle biogenesis
possibility that the closed conformation of STING dimer/CDNs is not at the Golgi complex. EMBO J 2012, 31:4535-4546.
essential for STING activation.
45. Hu MM, Yang Q, Xie XQ, Liao CY, Lin H, Liu TT, Yin L, Shu HB:
35. Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, Montealegre Sumoylation promotes the stability of the DNA sensor cGAS
Sanchez GA, Tenbrock K, Wittkowski H, Jones OY, Kuehn HS and the adaptor STING to regulate the kinetics of response to
et al.: Activated STING in a vascular and pulmonary syndrome. DNA virus. Immunity 2016, 45:555-569.
N Engl J Med 2014, 371:507-518.
46. Gonugunta VK, Sakai T, Pokatayev V, Yang K, Wu J, Dobbs N,
36. Jeremiah N, Neven B, Gentili M, Callebaut I, Maschalidi S, Yan N: Trafficking-mediated STING degradation requires
Stolzenberg MC, Goudin N, Frémond ML, Nitschke P, Molina TJ sorting to acidified endolysosomes and can be targeted to
et al.: Inherited STING-activating mutation underlies a familial enhance anti-tumor response. Cell Rep 2017, 21:3234-3242.
inflammatory syndrome with lupus-like manifestations. J Clin
Invest 2014, 124:5516-5520. 47. Prabakaran T, Bodda C, Krapp C, Zhang BC, Christensen MH,
Sun C, Reinert L, Cai Y, Jensen SB, Skouboe MK et al.:
37. Munoz J, Rodière M, Jeremiah N, Rieux-Laucat F, Oojageer A, Attenuation of cGAS-STING signaling is mediated by a p62/
Rice GI, Rozenberg F, Crow YJ, Bessis D: Stimulator of SQSTM1-dependent autophagy pathway activated by TBK1.
interferon genes-associated vasculopathy with onset in EMBO J 2018, 37.
infancy: a mimic of childhood granulomatosis with
48. Kagan JC, Medzhitov R: Phosphoinositide-mediated adaptor
polyangiitis. JAMA Dermatol 2015, 151:872-877.
recruitment controls Toll-like receptor signaling. Cell 2006,
38. Melki I, Rose Y, Uggenti C, Van Eyck L, Frémond ML, 125:943-955.
Kitabayashi N, Rice GI, Jenkinson EM, Boulai A, Jeremiah N et al.: 49. Kagan JC, Su T, Horng T, Chow A, Akira S, Medzhitov R: TRAM
Disease-associated mutations identify a novel region in couples endocytosis of Toll-like receptor 4 to the induction of
human STING necessary for the control of type I interferon interferon-beta. Nat Immunol 2008, 9:361-368.
signaling. J Allergy Clin Immunol 2017, 140:543-552.e5.
50. Husebye H, Aune MH, Stenvik J, Samstad E, Skjeldal F, Halaas O,
39. Konno H, Chinn IK, Hong D, Orange JS, Lupski JR, Mendoza A, Nilsen NJ, Stenmark H, Latz E, Lien E et al.: The Rab11a GTPase
Pedroza LA, Barber GN: Pro-inflammation associated with a controls Toll-like receptor 4-induced activation of interferon
gain-of-function mutation (R284S) in the innate immune regulatory factor-3 on phagosomes. Immunity 2010, 33:583-
sensor STING. Cell Rep 2018, 23:1112-1123. 596.
40. Tang ED, Wang CY: Single amino acid change in STING leads to 51. Yurchenko M, Skjesol A, Ryan L, Richard GM, Kandasamy RK,
constitutive active signaling. PLoS One 2015, 10:e0120090. Wang N, Terhorst C, Husebye H, Espevik T: SLAMF1 is required
for TLR4-mediated TRAM-TRIF-dependent signaling in human
41. Haag SM, Gulen MF, Reymond L, Gibelin A, Abrami L, Decout A, macrophages. J Cell Biol 2018, 217:1411-1429.
 Heymann M, van der Goot FG, Turcatti G, Behrendt R, Ablasser A:
Targeting STING with covalent small-molecule inhibitors. 52. Yu S, Nie Y, Knowles B, Sakamori R, Stypulkowski E, Patel C,
Nature 2018, 559:269-273. Das S, Douard V, Ferraris RP, Bonder EM et al.: TLR sorting by
This study identifies small-molecule STING antagonists that inhibit STING Rab11 endosomes maintains intestinal epithelial-microbial
palmitoylation by targeting Cys91. The authors show that these small- homeostasis. EMBO J 2014, 33:1882-1895.
molecule antagonists attenuate pathological features of autoinflamma-
tory disease in mice. Thus, as discussed by Mukai et al. (see Ref. [23]), 53. Akbar MA, Mandraju R, Tracy C, Hu W, Pasare C, Krämer H: ARC
STING palmitoylation is a therapeutic target for autoinflammatory dis-  syndrome-linked Vps33B protein is required for inflammatory
eases that are caused by aberrant cytosolic dsDNAs. endosomal maturation and signal termination. Immunity 2016,
45:267-279.
42. Hansen AL, Buchan GJ, Rühl M, Mukai K, Salvatore SR, Ogawa E, This study shows that Drosophila and mammalian Vps33B proteins play
 Andersen SD, Iversen MB, Thielke AL, Gunderstofte C et al.: Nitro- critical roles in the degradation of activated TLR4. Exaggerated inflam-
fatty acids are formed in response to virus infection and are matory responses dictated by persistence of TRL4 in aberrant endosomal
potent inhibitors of STING palmitoylation and signaling. Proc compartments could, therefore, contribute to symptoms of ARC syn-
Natl Acad Sci U S A 2018, 115:E7768-E7775. drome, a disease linked to loss of Vps33B.
This study identifies endogenously formed nitro-fatty acids that inhibit
STING palmitoylation by targeting Cys88/91. See annotations to Ref. [41]. 54. Cullinane AR, Straatman-Iwanowska A, Seo JK, Ko JS, Song KS,
Gizewska M, Gruszfeld D, Gliwicz D, Tuysuz B, Erdemir G et al.:
43. Linder ME, Deschenes RJ: Palmitoylation: policing protein Molecular investigations to improve diagnostic accuracy in
stability and traffic. Nat Rev Mol Cell Biol 2007, 8:74-84. patients with ARC syndrome. Hum Mutat 2009, 30:E330-7.

www.sciencedirect.com Current Opinion in Cell Biology 2019, 59:1–7

Descargado para Anonymous User (n/a) en Universidad Simon Bolivar de ClinicalKey.es por Elsevier en abril 19, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.

You might also like