You are on page 1of 11

REVIEWS

STING: infection, inflammation


and cancer
Glen N.Barber
Abstract | The rapid detection of microbial agents is essential for the effective initiation of
host defence mechanisms against infection. Understanding how cells detect cytosolic DNA
to trigger innate immune gene transcription has important implications not only for
comprehending the immune response to pathogens but also for elucidating the causes of
autoinflammatory disease involving the sensing of self-DNA and the generation of effective
antitumour adaptive immunity. The discovery of the STING (stimulator of interferon
genes)-controlled innate immune pathway, which mediates cytosolic DNA-induced
signalling events, has recently provided important insights into these processes, opening
the way for the development of novel immunization regimes, as well as therapies to treat
autoinflammatory disease and cancer.

Stimulator of interferon genes (STING; also known as and cancer. We also discuss new opportunities for the
Stimulator of interferon
genes
MITA and MPYS, and encoded by TMEM173) is a development of novel anti-inflammatory and anti
(STING). An endoplasmic signalling molecule associated with the endoplasmic tumour compounds that specifically target this key
reticulum-associated reticulum (ER) and is essential for controlling the signalling pathway.
protein that binds to cyclic transcription of numerous host defence genes, includ-
dinucleotides or cytosolic DNA
ing typeI interferons (IFNs) and pro-inflammatory The STING signalling pathway
to trigger cytokine production.
cytokines, following the recognition of aberrant DNA Activation of STING. STING is a 379 amino acid pro-
Cyclic dinucleotides species or cyclic dinucleotides (CDNs) in the cytosol tein, consisting of several transmembrane regions,
(CDNs). Secondary of the cell13. The sources of DNA that induce CDNs that is expressed in various endothelial and epithelial
messengers secreted by include the genome of invading pathogens, such as cell types, as well as in haematopoietic cells, such as
certain bacteria, such as
Listeria monocytogenes, or
herpes simplex virus 1 (HSV1), whereas certain bacte- Tcells, macrophages and dendritic cells (DCs), includ-
generated by cyclic GMPAMP ria can secrete CDNs following infection of the host 2,4. ing plasmacytoid dendritic cells (pDCs)1,2. Homologues
synthase (cGAS) following Recent reports have indicated that potent activators of of STING have been identified in different eukaryotic
binding to cytosolic DNA. the STING pathway may also include self-DNA that species as well as in invertebrates (BOX 2) . Early
has leaked from the nucleus of the host cell, perhaps studies showed that STING stimulates the transcrip-
following cell division or as a consequence of DNA tion of numerous innate immune genes in response to
damage5. Such DNA species may be responsible for invading DNA viruses, certain bacteria or transfected
causing various autoinflammatory diseases, such as DNA1,2,8,9. Further investigations clarified that STING
systemic lupus erythematosus (SLE) or AicardiGoutires is robustly activated by CDNs such as cyclic diAMP
syndrome (AGS), and may influence inflammation- (cdiAMP), which can be secreted by bacteria like
associated cancer 5. Polymorphisms in TMEM173 Listeria monocytogenes3,10.
have been reported to influence STING-dependent Cytosolic DNA species can trigger STING signalling
innate immune signalling and might affect suscepti- following binding to a 522 amino acid protein termed
bility to pathogen infection and contribute to severe cyclic GMPAMP synthase (cGAS) (FIG.1). In the presence
Department of Cell Biology inflammatory disorders (BOX1). of ATP and GTP, cGAS catalyses the production of a
and Sylvester Comprehensive STING signalling has now been shown to be essen- type of CDN referred to as cGAMP (cyclic GMPAMP),
Cancer Center, University of tial for protecting the cell against a variety of patho- which contains one 2,5-phosphodiester linkage and a
Miami School of Medicine, gens and even against the development of cancer by canonical 3,5 linkage (c[G(2,5)pA(3,5)p])1113. STING
Miami, Florida 33136, USA.
e-mail:
promoting antitumour immune responses6,7. Here, we is also known to bind double-stranded DNA (dsDNA)
gbarber@med.miami.edu review recent insights into the regulation of STING directly 14, although the physiological relevance of this
doi:10.1038/nri3921 signalling and its role in autoinflammatory diseases remains to be clarified.

760 | DECEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Lessons from STING variants


Subtle variations in the amino acid sequence of TMEM173 which encodes stimulator of interferon genes (STING)
have been shown to influence STING-dependent innate immune signalling. For example, human STING harbouring
an arginine at position 232 (R232) can be activated by bacterial cyclic dinucleotides (CDNs), such as cyclic di-AMP
(cdiAMP), as well as cyclic GMPAMP synthase (cGAS)-generated cyclic GMPAMP (cGAMP), whereas STING variants
with a histidine at this position (R232H) are poorly responsive to bacterial CDNs and are only effectively activated by
cGAMP97.The R293Q variant also only efficiently responds to cGAMP. Similar studies have indicated that, although the
human R71HG230AR293Q (HAQ) variant of STING is activated by all CDN types, it has a reduced ability to stimulate
typeI interferon (IFN) production compared with wild-type STING when overexpressed in 293T cells; the reason for this
remains unclear97,98. In mice, a T596A STING variant is also defective in inducing typeI IFN production in response to
CDNs4. Thus, certain single nucleotide polymorphisms (SNPs) within TMEM173 may predict susceptibility to pathogen
infection and perhaps the response to immunization regimes.
Of importance, several typeI IFN-inducing compounds, such as 10carboxymethyl9acridanone (CMA) and
5,6dimethylxanthenone4acetic acid (DMXAA), have been found to function by binding to and triggering STING
activity in mice, similarly to CDNs86,99. However, these compounds which attracted additional interest through their
considerable anticancer activity were not found to be effective drugs in humans owing to differences in structure
of the human STING protein86,97,99.
Alternative polymorphisms in the human TMEM173 gene have been reported to result in a gainoffunction
phenotype and are likely to contribute to severe inflammatory disorders. For example, patients suffering from vascular
and pulmonary syndrome (VAPS), which is typified by lesions in the cheeks, nose, fingers and toes, were found to have
mutations that result in STING hyperactivity100. Patients with VAPS were found to exhibit three mutations in exon 5 of
TMEM173 (N154S, V155M and V147L). These variants potently stimulated the typeI IFN promoter, as determined by
invitro expression studies in 293T cells100. Speculatively, these mutations may expedite STING trafficking from the
endoplasmic reticulum to the perinuclear region to stimulate cytokine production or may affect STING protein
stability, thereby sustaining STING activity101.
It is also not yet clear why patients with STING-associated vasculopathy with onset in infancy (referred to as SAVI)
which is an autoinflammatory disease caused by genetic mutations that result in STING hyperactivity, even in the
absence of cytosolic DNA species develop the disease. It is possible that STING-induced cytokines may affect
Systemic lupus endothelial function and aggravate macrophage activity in blood vessels5. Janus kinase (JAK) inhibitors, which prevent
erythematosus typeI IFN signalling, suppressed the induction of IFN-inducible genes in Tcells acquired from patients with SAVI100.
(SLE). An autoimmune disease Thus, such drugs may be useful for the treatment of patients with SAVI. It is tempting to consider whether mutations in
in which autoantibodies TMEM173 or in genes encoding proteins that directly or indirectly regulate STING, such as cGAS, may contribute to
specific for DNA, RNA or other types of autoinflammatory diseases102.
proteins associated with
nucleic acids form immune
complexes that damage small
blood vessels, especially in cGAS is a member of the nucleotidyltransferase fam- expressed7,18. The STING pathway is also independent
the kidneys. Patients with ily that includes the human dsRNA sensor oligoadenylate of AIM2 (absent in melanoma 2), which also inter-
SLE generally have abnormal
synthetase 1 (OAS1). Sequences of non-specific dsDNA acts with non-specific dsDNA species but triggers
B and Tcell function. SLE
can be associated with species greater than 30bp have been reported to stim- caspase 1mediated cleavage of the pro-inflammatory
hyperproduction of typeI ulate cGAS activity, with a single CDN generated by cytokines interleukin1 (IL1) and IL18 from their
interferons. cGAS binding to two molecules of STING in the ER15. precursor proteins19. However, STING signalling is
This event probably influences changes in STING con- known to potently influence the expression of the
AicardiGoutires
syndrome
formation, which leads to a striking trafficking event precursor proteins (but not their processing), and
(AGS). A neurodegenerative in which STING, complexed with TANK-binding therefore it is possible that STING may act in concert
disorder that can be caused kinase1 (TBK1), relocates to perinuclear regions of with the AIM2 pathway 20. Finally, although cGAS has
by STING-dependent cytokine the cell2,16. This process is required to deliver TBK1 to been clearly implicated in regulating STING func-
hyperproduction owing to
endolysosomal compartments where it phosphorylates tion through the generation of CDNs, other dsDNA-
mutations in genes such as
TREX1 (three-prime repair the transcription factors interferon regulatory factor 3 binding proteins have been shown to influence STING
exonuclease 1). (IRF3) and nuclear factor-B (NFB). These transcrip- activity, such as -IFN-inducible protein 16 (IFI16)
tion factors then translocate into the nucleus to initi- and DEAD box protein 41 (DDX41), although the
Cyclic GMPAMP synthase ate innate immune gene transcription (FIG.1). STING is underlying mechanisms remain to be clarified7.
(cGAS). A cellular protein
that binds to cytosolic DNA
then rapidly degraded, an event that may avoid problems
species and generates associated with sustained cytokine production17. Trafficking and regulation of STING. After binding to
stimulator of interferon genes STING signalling appears to be independent CDNs, STING (complexed with TBK1) traffics to peri
(STING)-activating cyclic of other DNA sensing pathways such as the Toll- nuclear regions via pre-autophagosomal-like structures;
dinucleotides.
like receptor 9 (TLR9) pathway, which is activated a process resembling autophagy (hereafter referred to
Oligoadenylate synthetase 1 by binding to unmethylated CpG dinucleotides of as STING-related autophagy) in which, generally under
(OAS1). A double-stranded approximately 21 bases long 18. Thus, the TLR9 and starvation conditions, cell organelles are engulfed in
RNA sensor that, in the STING signalling pathways have evolved to sense dif- a membraned vesicle (that is, an autophagosome) for
presence of ATP, generates ferent types of DNA species, although both pathways fusion with endosomes and lysosomes, which degrade
2,5linked oligoadenylates,
which can activate
use IRF3 and NFB to predominantly control gene and recycle the molecular components 2,16. These
RNaseLmediated viral RNA induction. Furthermore, TLR9 is mainly expressed pre-autophagosomal-like structures probably evolve
degradation. in pDCs and Bcells, whereas STING is more broadly from the ER, where STING is located1,21.

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | DECEMBER 2015 | 761

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 2 | The evolution of STING


or by mammalian target of rapamycin (mTOR), which
are switched on under conditions of cellular stress25.
In eukaryotes, no homologues of stimulator of interferon genes (STING) and Cytosolic dsDNA and/or CDNs were found to activate
cyclic GMPAMP synthase (cGAS) have been reported in plants or fungi, although the ULK12 axis and to initiate a negative feedback
homologues have been found in the choanoflagellate Monosiga brevicollis and in loop that controlled STING overactivity 17. Evaluation
cnidarians, such as sea anemone and Hydra magnipapillata103,104. Interestingly, the
of ULK12deficient mice may shed further light on
two molecules appear to have been lost in nematodes but reemerge in a variety of
insects such as Drosophila melanogaster. However, invertebrate cGAS homologues
the relationship of these kinases and STING func-
lack zinc ribbon domains probably required for DNA binding and have short tion. The significance of STING phosphorylation at
aminoterminal regions, which suggest they cannot bind DNA and/or generate additional serine sites is under evaluation, but they
cyclic dinucleotides (CDNs). By contrast, examination of STING sequences in are thought to have a role in the control of STING
invertebrates demonstrates that several key residues that are required for binding degradation17. Furthermore, several studies have now
CDNs are conserved. Thus, STING may have evolved to detect CDNs and/or nucleic indicated that STING is ubiquitylated on a number of
acid before vertebrate proteins such as cGAS evolved to synthesize CDNs such lysine residues26.
as cGAMP, although additional biochemical and biophysical evaluation is required In addition, the function of STING may be controlled
to confirm this. by microRNAs (miRNAs); a screen designed to identify
It remains to be determined whether STING exhibits an anti-pathogen function in
miRNAs that influence host defence to viral infection
invertebrates, similar to its function in mammalian cells. Studies have indicated that
human STING has evolved into a number of forms, each with a different single
identified miR5763p as a suppressor of STING27.
nucleotide polymorphism (SNP). For example, up to 20.4% of the population express Indeed, dsDNA- and CDN-triggered STING signalling
a form of TMEM173 (which encodes STING) with a triple R71HG230AR293Q (HAQ) was found to initiate miR5763p expression to prompt
SNP, while 13.7% have an R232H substitution, 5.2% have a G230AR293Q (AQ) a feedback mechanism that helps to prevent sustained
substitution and 1.5% have a R293Q substitution98,105. These SNPs appear to influence cytokine production.
STING function and may help to explain susceptibility to certain diseases (see BOX1). While the post-translational modifications that con-
trol STING function remain to be fully determined,
understanding these processes may help to explain
Studies indicate that STING forms part of the mechanisms of innate immune signalling events and
ERassociated translocon-associated protein (TRAP) perhaps autoinflammatory disease, as well as provide
complex, although it is not clear whether this structure potentially new therapeutic targets for drug intervention.
is required for the autophagy-like signalling process1.
Proteins intended for Nlinked glycosylation and/or STING signalling triggered by microorganisms
secretion are shunted through the TRAP complex into STING promotes immunity to DNA viruses and retro
the luminal region of the ER following translation, viruses. Many DNA viruses have been reported to
although it is unknown whether STING is essential trigger STING-dependent activity, including adenovirus,
for any of these activities17,22. This form of autophagy- vaccinia virus and papilloma virus2,2831.The importance
assisted typeI IFN production has been reported to of the cGASSTING axis has also been demonstrated
occur in pDCs that engulf DNA-containing immune for retrovirus infection including HIV3235. It is probable
complexes, an event noted to require TLR9 (REF.23). that retroviral DNA activates the cGASSTING axis,
Thus, both TLR9- and STING-mediated signalling may although a recent report indicated that retroviral RNA
similarly share an autophagy-related mechanism to DNA hybrids can also trigger this pathway suggest-
induce innate immune gene activation. ing that a wide array of nucleic acid structures may be
capable of triggering STING activity 32,33,3638. CDNs may
Translocon-associated
Post-translational modifications of STING. Attempts to also be packaged within viruses to directly stimulate
protein understand the mechanisms of STING signalling indi- STING antiviral signalling 39,40.
(TRAP). A complex of proteins cated that, in the presence of dsDNA, human STING is Recently, endogenous retroviruses (ERVs) have been
that shunt proteins destined rapidly phosphorylated on a number of serine residues reported to have a role in stimulating T cell-independent
for Nlinked glycosylation and
located at the carboxyterminal section. In one study, Bcell activity in response to exposure to bacterial
secretion into the endoplasmic
reticulum after translation. phosphorylation of S366 was found to inhibit the ability capsular polysaccharides or viral capsids41. This Bcell
of STING to activate IRF3, which largely prevented the response resulted from activation of both retinoic
Immune complexes induction of pro-inflammatory cytokines17. However, acid-inducible gene I (RIGI) and STING signalling
Complexes of antigen (including NFB signalling was not affected. This event may ensure pathways by reactivated ERVs. It remains unclear how
DNA) bound to antibody and,
sometimes, components of the
that pro-inflammatory protein production is inhibited the genomes of DNA viruses, such as HIV and HSV1,
complement system. The levels while ensuring that NFBdependent genes continue trigger STING signalling, since the capsids are thought
of immune complexes are to be expressed, perhaps to influence cell survival. to deliver the viral genome to the nucleus through the
increased in many autoimmune Screening assays designed to identify kinases involved nuclear pore33; however, it is plausible that the viral
disorders, in which they
in the regulation of STING by post-translational nucleic acids may be exposed from the protective capsids
become deposited in tissues
and cause tissue damage. modification indicated that the autophagy-related soon after entry into the cell or at the nuclear pore. It is
serine/threonine protein kinases ULK1 and ULK2 also possible that some DNA viruses, such as polyoma
MicroRNAs (referred to here as ULK12) can specifically phos- virus, replicate in the ER where STING resides42, which
(miRNAs). Small RNA phorylate S366 and help prevent chronic STING may help hide the viral genome from cytosolic or
molecules that regulate the
expression of genes by binding
activity 17. TBK1 has also been suggested to target this nuclear sensors, such as cGAS. It thus remains to be fully
to the 3 untranslated regions site to control STING24. The function of ULK12 can determined how STING signalling is triggered through
(3 UTR) of specific mRNAs. be regulated by AMP-activated protein kinase (AMPK) cGAS by pathogen-associated DNA in thecell.

762 | DECEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

DNA virus Type I IFNs

Type I IFN
receptor

Mitochondrion
Viral
DNA
P P
cGAS JAK TYK

dsDNA
Self-DNA leaked STAT1
from nucleus ISGF3 STAT2
Intracellular
GTP bacteria IRF9
and ATP
CDNs
cGAMP c-di-AMP

Endolysosome
STING
Autophagy
p50 p65 IRF3 TBK1
NF-B

ER
P
p50 p65 IRF3
Perinuclear Nucleus
NF-B Golgi
NF-B
Type I
IRF3 p50 p65
IFNs

IFN-stimulated
genes

Figure 1 | STING-dependent innate immune signalling. Stimulator of interferon genes (STING) is activated by cyclic
dinucleotides (CDNs) produced by certain bacteria or by cyclic GMPAMP synthase (cGAS),Nature which in the presence
Reviews of ATP
| Immunology
and GTP catalyses the production of a type of CDN referred to as cGAMP (cyclic GMPAMP) following binding to cytosolic
DNA species (from viruses or bacteria, or self -DNA from the nucleus or mitochondria). STING is associated with the
endoplasmic reticulum (ER) and, following binding to CDNs, STING forms a complex with TANK-binding kinase 1 (TBK1).
This complex traffics to the perinuclear Golgi via pre-autophagosomal-like structures a process resembling autophagy
to deliver TBK1 to endolysosomal compartments where it phosphorylates the transcription factors interferon regulatory
factor 3 (IRF3) and nuclear factor-B (NFB). Stimulation of the IRF3 and NFB signalling pathways leads to the induction
of cytokines and proteins, such as the typeI interferons (IFNs), that exert anti-pathogen activity. c-di-AMP, cyclic di-AMP;
dsDNA, double-stranded DNA; ISGF3, interferon-stimulated gene factor3; JAK, Janus kinase; STAT, signal transducer and
activator of transcription; TYK, tyrosine kinase.

STING suppresses replication of RNA viruses. It The synthase cGAS has also been shown to play a part
became evident soon after its discovery that STING is in host defence against RNA virus infection, although
also required for host protection against a number of the mechanisms remain to be clarified. For example,
RNA-related pathogens, including vesicular stomatitis ablation of this enzyme rendered RNA virus-infected
virus, Sendai virus and possibly dengue virus 1,2,43. cells sensitive to viral replication, even though RNA
However, synthetic RNA, such as polyinosinic species do not trigger cGAS activity or the generation of
polycytidylic acid (poly(I:C)), or RNA viruses do not CDNs44,45. It is therefore possible that STING may have
trigger STING-related autophagy or the innate immune evolved two disparate host defence-related functions
signalling events that usually result in gene induction1,2. depending on whether the invading pathogen possessed
Thus, the mechanisms by which STING thwarts gene an RNA or DNA genome. DNA viruses may trigger
replication of positive- and negative-sense viruses needs potent STING-activated gene induction, whereas RNA
to be clarified. It is possible that STING exerts this virus replication may be thwarted at the level of trans
effect through its translocon function, conceivably by lation or post-translation. Alternatively, it is possible that
regulating translation of viral RNA or even autophagy. STING signalling may be responsible for constitutively

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | DECEMBER 2015 | 763

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Viral to be triggered through the STING pathway by several


antigen bacteria including Legionella pneumophila, Francisella
Cross-
MHC class I presentation tularensis, Chlamydia muridarum, Streptococcus pyogenes,
Brucella abortus and Mycobacterium tuberculosis5055.
Although the activation of innate immune genes
through STING signalling may help prevent dangerous
DNA virus Phagocyte bacterial infection (FIG.2), in certain situations STING-
mediated innate immune gene induction surprisingly
Pro-inammatory appears to facilitate bacterial survival. For example,
CDNs from genes L. monocytogenes and M. tuberculosis may activate
intracellular STING to downregulate cell-mediated immunity and host
cGAS bacteria Phagocyte
engulfment defence responses55. Patients with severe tuberculosis are
known to present with high IFN levels, which correspond
to disease severity. Plausibly, typeI IFN-induced genes
STING Cytokine may comprise cytokines that suppress adaptive immune
receptor responses, perhaps in an effort to control inflammation.
Type I IFNs Of note, mice deficient for both the TLR adaptor molecule
Pro-inammatory MYD88 and STING retained the ability to mount
cytokines adaptive immune responses to L.monocytogenes, sug-
IFN
Nucleus Type I or IFN
gesting that alternative sensing pathways can recognize
IFN receptor bacterial infection and trigger adaptive immunity in the
absence of STING55. This could involve activation of
Infected cell
the RIGIlike pathway by bacterialRNAs.
These studies highlight the delicate equilibrium
between an appropriate immune response and inflam-
Anti-pathogen mation, a balance that has perhaps been exploited by
gene induction microorganisms. Such findings may have important
implications in the development of STING-targeting
adjuvants and the design of vaccines intended to induce
Uninfected cell robust, long-lasting adaptive immune responses (FIG.2).

Protection of STING and autoinflammatory disease


uninfected cell
Although transient inflammatory responses are essen-
Figure 2 | STING signalling is essential for anti-pathogen host defence. DNA tial for initiating host defence responses to pathogen
viruses and cyclic dinucleotides (CDNs) produced by certain bacteria stimulate invasion, sustained or chronic inflammatory signalling
stimulator of interferon genes (STING) activity to trigger the production
Nature of |primary
Reviews Immunology is a key factor in the development of autoimmune dis-
innate immune genes. The products from these genes can exert direct intracellular
ease. Over 20% of individuals in the United States alone
anti-pathogen activity as well as trigger the production of typeI interferons (IFNs) and
other cytokines that can protect uninfected cells and stimulate adaptive immune
will suffer some form of inflammatory disease, such as
responses. cGAS,cyclic GMPAMP synthase. rheumatoid arthritis (RA), SLE or inflammatory bowel
disease56. In almost all cases, the aetiology of such dis-
eases remains unknown. What is known is that patients
generating low levels of typeI IFNs that are required for with complex inflammation-related disorders, such as
the robust activation of RNA-sensing pathways follow- RA and SLE, exhibit high levels of antinuclear antibody
ing viral infection. Perhaps unsurprisingly, some recent (ANA), DNA and even nucleosomes in the blood, as well
studies have reported that some viruses can suppress as high levels of circulating cytokines57. Self-DNA, prob-
STING function. For example, the non-structural ably from necrotic or inappropriately apoptosed cells,
protein 2B (NS2B)NS3 protease complex of dengue has therefore been implicated in the pathogenesis of
virus has been shown to cleave human STING, and these types of disease, with aberrant innate immune sig-
severe acute respiratory syndrome (SARS) coronavirus nalling being caused by a failure to distinguish between
reportedly inhibits the ability of STING to activate IRF3 self and foreign nucleicacid58.
(REF.46). Furthermore, adenovirus and HSV1 as well as The inability to efficiently eliminate self-DNA that
other viruses have similarly been described to control could inadvertently trigger immune responses may also
STING function1,2,4749. be a key factor in facilitating inflammatory disease. For
example, a number of animal models with defects in
The STING pathway and bacteria. Gram-negative and DNases exhibit lethal inflammatory disorders. In some
Gram-positive bacteria have also been reported to pro- instances, DNase-related defects have been found in
mote STING signalling 1,2. For example, L.monocytogenes humans with comparable disease57. Defects in DNASE1,
does this by secreting STING-activating CDNs (that is, for example, have been implicated in autoimmune dis-
cdiAMP), although the genomes of bacteria have also orders in humans and mice59. Furthermore, mice with
been shown to be capable of stimulating STING func- defects in the lysosomal DNA endonuclease Dnase2,
tion14,10. TypeI IFN production has now been shown which is essential for degrading DNA from apoptotic

764 | DECEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

knockout mice were relatively disease free, exhibiting


reduced cytokine levels, reduced ANA levels and no
Self-DNA signs of inflammatory myocarditis or early death62. Bone
marrow-derived macrophages (BMDMs) or bone marrow-
derived dendritic cells (BMDCs) from Trex1/ mice,
cGAS but not from Trex1/Tmem173/ mice, were found to
TREX1 produce high levels of inflammatory cytokines in the
No degradation CDN absence of any notable infection5,62,64. Furthermore,
of self-DNA the adoptive transfer of wild-type bone marrow to
Constitutive irradiated Trex1/ mice rescued mortality 62. Thus,
STING activation
STING-dependent inflammatory cytokines produced
by immune cells probably have an important role in
Additional immune TREX1mediated inflammatory disease65 (FIG.3).
Hyperproduction
of cytokines and cell recruitment However, it has been reported that Trex1/ cells dis-
chemokines and activation play a defect in the transition from G1 phase to S phase
Macrophage
of the cell cycle and also accumulate DNA species in the
cytoplasm63. BMDCs from Trex1/ mice in G2 arrest
exhibited increased levels of cytokine expression com-
pared with BMDCs from wild-type mice, suggesting
that TREX1 may have a housekeeping role in eliminat-
Autoinammation ing aberrant self-DNA species that may accrue in the
cytoplasm after genome replication62,63. These nucleic
Figure 3 | STING in autoinflammatory disease. Cyclic GMPAMP synthase (cGAS) binds acids may intrinsically activate cytokine production
to cytosolic self-DNA and generates stimulator of interferon Nature Reviews | Immunology
genes (STING)-activating through activation of cGAS and STING. A further
cyclic dinucleotides (CDNs). The activation of STING by self-DNA can induce
report indicated that DNA damaged by ultraviolet light
autoinflammatory disease. Mutations in three-prime repair exonuclease 1 (Trex1), which
encodes a DNA exonuclease that degrades single-stranded DNA and double-stranded
and harbouring oxidative modifications was unable to
DNA substrates, can cause STING activation through failure to eliminate self-DNA that has be degraded by TREX1 (REF.66). Such DNA could trigger
leaked into the cytosol of the cell. Conversely, mutations in STING itself can lead to STING activity and may constitute a new damage-
constitutive activity and autoinflammtory diseases such as STING-associated vasculopathy associated molecular pattern (DAMP) that can aggravate
with onset in infancy. STING-dependent autoinflammatory disease.
Collectively, these studies show that the body has
devised several mechanisms to prevent self-DNA from
cells engulfed by phagocytes, die before birth owing activating host defence-related DNA sensors, such as
to the toxic effects of typeI IFN overproduction 57. STING. TREX1 probably prevents intrinsic self-DNA-
However, Dnase2/ mice are viable when crossed with mediated activation of STING signalling, whereas
mice deficient for STING (Tmem173/ mice), as the pro- DNase II may prevent STING-dependent activation by
duction of apoptotic DNA-driven cytokine production extrinsic signals in phagocytes. These DNases constitute
by phagocytes is almost completely eliminated20. Mice indirect negative regulators of STING and, when defec-
defective in three-prime repair exonuclease 1 (Trex1; also tive, they can result in constitutive STING activity and
known as Dnase3) similarly exhibit significant cytokine the onset of inflammatory disease (FIG.3). Understanding
overproduction and inflammation and, importantly, these processes may enable the design of new drugs for
TREX1related mutations with concomitant disease the treatment of inflammatory disease.
have been reported in humans60. This indicates that
STING signalling is responsible for the production of Mitochondrial DNA and STING
harmful pro-inflammatory cytokines generated by self- As discussed, the STING signalling pathway probably
DNA derived from necrotic or inappropriately apop- arose to detect aberrant cytosolic DNA species from an
tosed cells. Such DNases almost certainly help ensure infectious agent or from leakage of self-DNA from the
Three-prime repair that apoptotic cells remain immunologically silent nucleus of damaged cells7. However, these might not be
exonuclease 1 following phagocytosis. the only sources of cytosolic DNA. The hundreds, if not
(TREX1). A DNase that
degrades self-DNA species
STING-dependent signalling was also found to be the thousands, of mitochondria in the cytosol of most cells
that can otherwise chronically cause of inflammation-aggravated lethality in Trex1/ contain 210 copies of mitochondrial DNA (mtDNA),
activate stimulator of mice61,62. TREX1 is a 3 to 5 DNA exonuclease that which exist as closed circular dsDNA species of approxi-
interferon genes (STING) degrades single-stranded DNA (ssDNA) and dsDNA mately 15,000 base pairs67. Recent evidence indicates that
signalling.
substrates. Trex1/ mice are viable but have a median mitochondrial stress, such as that induced by viruses, can
Damage-associated lifespan of approximately 10weeks owing, in part, to the cause mtDNA leakage into the cytosol that can activate
molecule pattern development of inflammatory myocarditis63. Patients with the STING pathway and the production of cytokines68.
(DAMP). A cellular molecule, severe SLE and AGS have been found to have mutations in Although this particular example may be of benefit to the
such as uric acid, ATP and TREX1, suggesting that defects in this protein are respon- host, the sensing of mtDNA that has leaked into the cyto-
heat shock proteins, that can
activate the inflammatory
sible for these diseases, possibly through the inability of sol following apoptosis-induced mitochondrial stress may
response in the absence of phagocytes to degrade self-DNA60. Such patients typically have detrimental effects. However, as up to 50 billion cells
infection. die at a young age. However, Trex1/Tmem173/ double undergo apoptosis in the body per day, it is thus inevitable

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | DECEMBER 2015 | 765

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

that the body has developed an effective means to enable phagocytic infiltration69,74,75. Indeed, mice deficient for
programmed cell death events to be immunologically MYD88 or for tumour necrosis factor (TNF) are rela-
silent. Indeed, certain caspases may prevent the induction tively resistant to DMBA-induced skin carcinogenesis,
of inflammatory responses by cytosolic mtDNA69,70. indicating the importance of MYD88dependent signal-
The process of apoptosis involves the activation of ling and induction of the pro-inflammatory cytokine
the pro-death molecules BAK and BAX, which induce TNF in this process74,75.
mitochondrial outer-membrane permeabilization (MOMP) The trigger that controls primary innate immune
and causes the release of cytochromec into the cyto- responses as a consequence of DNA damage, however,
sol where it forms a complex with apoptotic protease- has remained largely unknown. Evidence indicates that
activating factor 1 (APAF1) and caspase 9, termed TLR-independent pathways could have a key role in this
the apoptosome. This in turn activates caspase 3 and process74. Furthermore, it has recently been shown that
caspase7, which start to target cellular proteins. Despite STING-deficient mice are resistant to DMBA-induced
the fact that certain caspases have an important role in skin polyp formation76. This study showed that DMBA-
apoptosis, they are not essential for cell death and invivo initiated DNA damage lead to nucleosome leakage into
clearance of cells, suggesting that they may exert other the cytosol and triggered STING-dependent cytokine pro-
important functions71. Indeed, recent studies have shown duction by the self-DNA. It is possible that DNA-damaged
that BAK- and BAX-induced MOMP can also cause keratinocytes intrinsically produce pro-inflammatory
the release of mtDNA, the induction of cGASSTING cytokines that attract phagocytes, which may facilitate the
signalling and typeI IFN production69,70. The effector removal of the damaged cells. These infiltrating immune
caspases, caspase 3, caspase 7 and caspase 9, were shown cells may engulf damaged keratinocytes, and the cellular
to be required to prevent mtDNA-induced STING acti- DNA from the dying cells may extrinsically activate
vation69,70. How this occurs is not yet fully apparent STING signalling to further propagate pro-inflammatory
but may involve caspase-mediated targeting of pro- cytokine production74 (FIG.4). This model is supported by
teins involved in STING-dependent signalling or even the observation that the adoptive transfer of wild-type
indirect targeting of mtDNA. Of course, such events bone marrow cells to Tmem173/ mice which are
require that STING be present in cells undergoing apop- resistant to DMBA-induced skin tumour development
tosis and fortunately, STING is not abundantly expressed results in the development of skin tumours similar to
in cells such as hepatocytes, which are known to contain those that develop in control mice following treatment
thousands of mitochondria7. A further conundrum is with DMBA62,76. Thus, STING signalling in haematopoietic
that caspase-deficient mice do not necessarily develop cells, similar to MYD88 signalling, plays an important
autoimmune disease, suggesting that caspase loss may part in facilitating DMBA-induced tumorigenesis.
affect other elements of the immune system to prevent STING-dependent dsDNA-induced innate immune
disease development 71. gene transcription is unaffected in Myd88/ cells, which
It is interesting to note that dysfunctional mito suggests that MYD88 activation occurs downstream
chondria have been reported to occur in autoimmune of STING induction in response to DNA-damaging
diseases such as SLE, but whether disease pathogenesis agents76. Thus, the current model suggests that DNA
involves release of mtDNA and activation of STING damage triggers STING-driven production of cytokines,
remains to be determined72. It has also been suggested which in an autocrine or paracrine manner can bind to
that inhibitors of effector caspases could serve as anti receptors that signal via the MYD88 adaptor molecule,
viral therapies because they may upregulate the such as the IL1 receptor and TLRs. This further drives
production of typeI IFNs by released mtDNA72. In the production of additional cytokines and growth
summary, the immunological silencing of programmed factors that propagate inflammation and facilitate skin
cell death involves an unclarified mechanism that pre- tumorigenesis. It is unclear whether STING has a role
vents leaked mtDNA from triggering STING-dependent in other types of inflammation-aggravated cancer, such
inflammatory processes. as hepatocellular carcinoma. Clearly, understanding
the involvement of STING in such events may lead to the
STING signalling in cancer design of new anticancer therapeutics.
Mitochondrial STING-aggravated tumorigenesis. Chronic inflammatory
outer-membrane signalling may not only instigate autoinflammatory disease Antitumour effects of STING. Although MYD88 sig-
permeabilization
(MOMP). Permeablization
such as AGS or STING-associated vasculopathy with onset nalling facilitates carcinogenesis in the skin, MYD88
induced by virus infection in infancy (SAVI) but can also contribute to the develop- signalling is protective in models of colitis-associated
and/or apoptosis to release ment of cancer, probably through cytokines, chemokines carcinogenesis (CAC), which can be experimentally
mitochondrial DNA that could and growth factors that stimulate cellular proliferation induced by carcinogens and inflammatory agents such
conceivably activate stimulator
and survival, as well as by promoting angiogenesis73. as azoxymethane (AOM) and dextran sulfate sodium
of interferon genes (STING).
Inflammation induced by carcinogens, which (DSS)74,76. It is possible that cytokines such as IL1 and
Angiogenesis cause DNA mutations, aggravates tumour develop- IL18 are secreted by damaged intestinal cells to facilitate
The development of ment by mechanisms that remained to be deter- wound repair 20,72,77,78. In the absence of wound repair,
new blood vessels from mined. In one example, the polyaromatic hydrocarbon concomitant inflammation alters the microbial
existing blood vessels. It is
frequently associated with
7,12dimethylbenz[]anthracene (DMBA) can drive the composition in the gut to species with enhanced geno-
tumour development and development of cutaneous skin tumours by promoting toxic capacity, resulting in further inflammation, DNA
inflammation. pro-inflammatory cytokine production and stimulating damage and tumorigenesis79,73.

766 | DECEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

The phenotype of STING-deficient mice treated STING-dependent cytokine production, including the
with AOM and DSS mimicked that of similarly treated production of IL1. It is thus possible that cGAS
MYD88deficient mice; the loss of STING rendered STING recognizes early DNA damaging events to trig-
mice susceptible to CAC 80. Further investigation ger the production of wound repair-initiating cytokines
showed that AOM induced DNA damage and triggered (FIG.4b). STING activation in damaged intestinal cells

a DMBA b AOM and DSS


IL-1R

IL-1
MYD88
DNA damage Cytokine DNA damage
production
Immune cell STING
recruitment

Nucleosome Cytokine
release? Cytosolic DNA from
production
phagocytosed cell
STING
STING
Cytosolic Intestinal
self-DNA Phagocyte epithelial cell

Keratinocyte
Engulfment
of damaged Propagation of IL-1 and
keratinocyte inammation IL-18
Tumour growth Wound repair

Antitumour
c immune response CTL

TCR
DNA damage Pro-inammatory Immune cell Tumour antigen Cross-priming
gene induction recruitment MHC class I

Nucleosome
release STING CDN

Type I IFNs cGAS

cGAS CDN STING


CD8+ DC
Dying tumour cell

Lysosome
Engulfment

Figure 4 | STING control of tumour development. a | Stimulator of and cause activation of the intrinsic STING pathway in intestinal cells. This
interferon genes (STING)-driven inflammation-associated cancer. event produces cytokines such as interleukin1 (IL1) or IL18 that enable
Carcinogens such as 7,12-dimethylbenz[]anthracene (DMBA) cause DNA wound repair to commence. Loss of STING Nature Reviews
prevents wound| Immunology
repair from
damage, which can result in the leakage of DNA into the cytosol and the occurring and may enable the infiltration and growth of genotoxic bacteria
intrinsic chronic activation of the STING pathway. This event attracts that trigger STING-independent inflammation and perhaps cancer (not
phagocytes that engulf damaged cells. The self-DNA from engulfed cells shown). c | STING-dependent antitumour cytotoxic T lymphocyte (CTL)
can extrinsically activate STING in the phagocytes, which results in the priming. Dying tumour cells are engulfed by antigen-presenting cells such
production of more cytokines and growth factors that can activate tumour as CD8+ dendritic cells (DCs). DNA from the engulfed cell triggers
growth-promoting pathways as well as attract more immune cells to the STING-dependent cytokine production in the phagocyte, which facilitates
region. b | STING and wound repair function. Carcinogens and inflammatory cross-presentation and antitumour CTL responses. Agonists of STING have
agents such as azoxymethane (AOM) and dextran sulfate sodium (DSS) also been shown to exert potent antitumour activity. CDN, cyclic dinucleotide;
cause DNA damage that can result in the leakage of DNA into the cytosol cGAS, cyclic GMPAMP synthase; IFN, interferon; TCR, Tcell receptor.

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | DECEMBER 2015 | 767

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

also activates typeI IFNs, which exert potent effects on to exert antitumour activity but was only specific for
the priming of antitumour Tcells (see later). Thus, loss mouse STING and failed to work in human patients86.
of STING may enable damaged cells to escape tumour However, several reports indicate that CDNs that bind
immunosurveillance8082. In summary, STING signal- human STING can exert antitumour activity in animal
ling may have an important protective effect against studies87. In some instances, STING agonists were shown
CAC, the second most common cancer in both men to be effective against tumours that were resistant to pro-
and women. Further analysis of the function of STING grammed cell death protein 1 (PD1) blockade88. Such
in controlling colorectal and/or intestinal wound agonists have also been shown to be experimentally useful
repair, as well as its influence on the composition of the as adjuvants in anticancer vaccine studies89,90. Thus, the
microbiota, will no doubt shed further insight into development of novel STING activators may lead to
the importance of STING in influencing tumorigenesis. the generation and rapid expansion of immunotherapy
trials to combatcancer.
STING signalling and adaptive antitumour immunity. Finally, it is noteworthy that anticancer drugs such
Adaptive Tcell responses are important for the control as cisplatin and etoposide are also DNA adduct-form-
and eradication of tumour cells82. Numerous immuno- ing agents that trigger cell death by instigating DNA
therapeutic strategies involving stimulating the adaptive damage91. These drugs were found to cause nuclear
immune response against cancers through checkpoint DNA leakage into the cytosol and to trigger intrinsic
blockade are presently under evaluation in clinical trials. STING-dependent cytokine activity 62. It is tempting
However, how adaptive immune responses are generated to speculate that such drugs may exert their anticancer
by DCs against tumour cells remains unknown. What is effects, in part, by stimulating the STING pathway and
apparent is that typeI IFN production by CD8+ DCs activating the immune system. Conversely, defects in
is involved in tumour antigen-specific Tcell activa- STING signalling may contribute to chemoresistance
tion through the cross-presentation of antigen and T cell in certain types ofcancer.
priming 83. The signalling pathway responsible for typeI The importance of STING in generating cytokines in
IFN induction is thought to be independent of the TLR response to DNA damage has been further emphasized
or RIGImitochondrial antiviral signalling (MAVS) in cells with defects in their DNA repair machinery 92,93.
protein pathways. Dysfunction in ataxia-telangiectasia mutated (ATM), a
Recent studies have shown that the STING pathway is kinase that facilitates DNA repair, or in the cobinding
essential for radiation-induced and spontaneous natural protein meiotic recombination 11 homologue (MRE11)
antitumour T cell responses 83,84. STING-deficient lead to the production of cytokines, which may con-
mice are unable to generate efficient antitumour Tcell tribute to the inflammatory phenotypes noted in
responses and prevent melanoma tumour growth. patients with Ataxia telangiectasia93. Loss of DNA repair
Furthermore, the ability of checkpoint inhibitors to mechanisms may enable self-DNA to leak out of the
stimulate Tcell responses was also abrogated in STING- cytoplasm to activate STING, or STING may be tar-
deficient mice, indicating a role for STING in the effi- geted by DNA repair machinery directly or indirectly
cacy of checkpoint inhibitors79. One hypothesis for to induce signalling 76,93. Collectively, it is becoming
the underlying mechanism is that CD8+ DCs engulf apparent that STING has a key role in facilitating anti-
Checkpoint blockade necrotic tumour cells, and the tumour cell-derived DNA tumour immune responses. Furthermore, stimulating
The targeting of Tcell triggers STING signalling in the DC20,8385. The resultant STING activity within the tumour microenvironment
coinhibitory receptors, such typeI IFNs, functioning in a paracrine or autocrine may comprise a new immunotherapeutic strategy to
as cytotoxic T lymphocyte
antigen4 (CTLA4) or
manner, may induce the production of additional help treat malignant disease.
programmed cell death proteins in the DC that facilitates cross-presentation and
protein1 (PD1), by antibodies Tcell activation (FIG.4c). Concluding remarks
that have clinical activity in It is known that apoptotic cells do not potently Growing evidence indicates that the intrinsic STING
multiple cancer types.
activate innate immune signalling. But, engulfment of pathway is crucial for recognizing DNA pathogens
Cross-presentation necrotic cells may trigger cytokine production, albeit or damaged DNA in the cytosol. The innate immune
The initiation of a CD8+ T cell at low levels, in a STING-dependent manner 20. Usually, response that ensues can attract immune cells that
response to an antigen DNase II in phagocytes efficiently digests engulfed cel- phagocytose the infected or damaged cells. The DNA
that is not present within lular DNA in the lysosomal compartment, and it is not from engulfed cells can stimulate extrinsic STING
antigen-presenting cells (APCs).
This exogenous antigen must
yet clear why some host DNA from necrotic cells escape signalling in the phagocytes, thereby promoting further
be taken up by APCs and then this process. It is possible that STING is activated by inflammatory signals.
rerouted to the MHC class I DNA that is present in autophagosomes or in engulfed The engulfment of apoptotic cells is immunologi-
pathway of antigen exosomes83,84. Collectively, further understanding of the cally silent, perhaps owing to the expression of caspases
presentation.
role of STING in facilitating adaptive immune responses and DNases that prevent intrinsic and extrinsic STING
Ataxia telangiectasia may have notable consequences in the design of future activation. However, cancer cells are able to stimulate
A neurodegenerative disease anticancer immunotherapies. modest extrinsic STING activity in antigen-presenting
caused by mutations in the Insight into the role of STING in facilitating anti cells such as CD8+ DCs, and this appears to be a suf-
ATM (ataxia-telangiectasia tumour T cell responses has stimulated interest in evalu- ficient and essential process for the efficient priming of
mutated) gene, the product
of which is essential for the
ating whether STING agonists could be useful therapies antitumour Tcells. Understanding the underlying mecha-
recognition and repair of to treat cancer. The STING activator 5,6dimethyl nisms of this cross-priming may lead to the development
DNA breaks. xanthenone4acetic acid (DMXAA) was already known of potent antitumour vaccines and therapies94,95.

768 | DECEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

It remains to be seen what other types of inflammatory types of cancer 83,96. Thus, designing drugs that trigger
or other diseases may be caused by defects in the STING or repress cGAS or STING activation and/or signalling
pathway. But it is possible that suppressing STING activity could be of interest to the anticancer, anti-pathogen,
may help to avoid autoinflammatory disease and certain anti-inflammatory and vaccine researchfields.

1. Ishikawa,H. & Barber,G.N. STING is an endoplasmic 21. Gomes,L.C. & Dikic,I. Autophagy in antimicrobial 45. Schoggins,J.W. etal. Pan-viral specificity of IFN-
reticulum adaptor that facilitates innate immune immunity. Mol. Cell 54, 224233 (2014). induced genes reveals new roles for cGAS in innate
signalling. Nature 455, 674678 (2008). 22. Nyathi,Y., Wilkinson,B.M. & Pool,M.R. immunity. Nature 505, 691695 (2014).
This is the first report on the discovery of STING Cotranslational targeting and translocation of 46. Maringer,K. & Fernandez-Sesma,A. Message in a
as an essential innate immune regulator. proteins to the endoplasmic reticulum. Biochim. bottle: lessons learned from antagonism of STING
2. Ishikawa,H., Ma,Z. & Barber,G.N. STING regulates Biophys. Acta 1833, 23922402 (2013). signalling during RNA virus infection. Cytokine Growth
intracellular DNA-mediated, typeI interferon- 23. Henault,J. etal. Noncanonical autophagy is required Factor Rev. 25, 669679 (2014).
dependent innate immunity. Nature 461, 788792 for typeI interferon secretion in response to DNA- 47. Kalamvoki,M. & Roizman,B. HSV1 degrades,
(2009). immune complexes. Immunity 37, 986997 (2012). stabilizes, requires, or is stung by STING depending
3. Burdette,D.L. etal. STING is a direct innate immune 24. Liu,S. etal. Phosphorylation of innate immune on ICP0, the US3 protein kinase, and cell derivation.
sensor of cyclic diGMP. Nature 478, 515518 (2011). adaptor proteins MAVS, STING, and TRIF induces Proc. Natl Acad. Sci. USA 111, E611E617
This report demonstrates that CDNs bind to and IRF3 activation. Science 347, aaa2630 (2015). (2014).
activate STING. 25. Alers,S., Loffler,A.S., Wesselborg,S. & Stork,B. 48. Zhou,Q. etal. The ERassociated protein ZDHHC1
4. Sauer,J.D. etal. The NethylNnitrosourea-induced Role of AMPK-mTORUlk1/2 in the regulation of is a positive regulator of DNA virus-triggered,
Goldenticket mouse mutant reveals an essential autophagy: cross talk, shortcuts, and feedbacks. MITA/STING-dependent innate immune signaling.
function of Sting in the invivo interferon response to Mol. Cell. Biol. 32, 211 (2012). Cell Host Microbe 16, 450461 (2014).
Listeria monocytogenes and cyclic dinucleotides. 26. Liu,X., Wang,Q., Pan,Y. & Wang,C. Sensing and 49. Luecke,S. & Paludan,S.R. Innate recognition of
Infect. Immun. 79, 688694 (2011). responding to cytosolic viruses invasions: An orchestra alphaherpesvirus DNA. Adv. Virus Res. 92, 63100
5. Ahn,J. & Barber,G.N. Self-DNA, STING-dependent of kaleidoscopic ubiquitinations. Cytokine Growth (2015).
signaling and the origins of autoinflammatory disease. Factor Rev. 26, 379387 (2015). 50. Mitzel,D.N., Lowry,V., Shirali,A.C., Liu,Y. &
Curr. Opin. Immunol. 31, 121126 (2014). 27. Yarbrough,M.L. etal. Primate-specific miR5763p Stout-Delgado,H.W. Age-enhanced endoplasmic
6. Woo,S.R., Corrales,L. & Gajewski,T.F. sets host defense signalling threshold. Nat. Commun. reticulum stress contributes to increased Atg9A
The STING pathway and the Tcell-inflamed tumor 5, 4963 (2014). inhibition of STING-mediated IFN- production
microenvironment. Trends Immunol. 36, 250256 28. Lam,E., Stein,S. & Falck-Pedersen,E. Adenovirus during Streptococcus pneumoniae infection.
(2015). detection by the cGAS/STING/TBK1 DNA sensing J.Immunol. 192, 42734283 (2014).
7. Barber,G.N. STING-dependent cytosolic DNA sensing cascade. J.Virol. 88, 974981 (2014). 51. Prantner,D., Darville,T. & Nagarajan,U.M.
pathways. Trends Immunol. 35, 8893 (2014). 29. Sunthamala,N. etal. E2 proteins of high risk human Stimulator of IFN gene is critical for induction of IFN-
8. Zhong,B. etal. The adaptor protein MITA links virus- papillomaviruses down-modulate STING and IFN- during Chlamydia muridarum infection. J.Immunol.
sensing receptors to IRF3 transcription factor transcription in keratinocytes. PLoS ONE 9, e91473 184, 25512560 (2010).
activation. Immunity 29, 538550 (2008). (2014). 52. Storek,K.M., Gertsvolf,N.A., Ohlson,M.B. &
9. Jin,L. etal. MPYS, a novel membrane tetraspanner, 30. Ablasser,A. etal. Cell intrinsic immunity spreads to Monack,D. M. cGAS and Ifi204 cooperate to produce
is associated with major histocompatibility complex bystander cells via the intercellular transfer of cGAMP. type I IFNs in response to Francisella infection.
class II and mediates transduction of apoptotic Nature 503, 530534 (2013). J.Immunol. 194, 32363245 (2015).
signals. Mol. Cell. Biol. 28, 50145026 (2008). 31. Lau,L., Gray,E.E., Brunette,R.L. & Stetson,D.B. 53. Watson,R.O., Manzanillo,P.S. & Cox,J.S.
10. Woodward,J.J., Iavarone,A.T. & Portnoy,D. A. DNA tumor virus oncogenes antagonize the cGAS- Extracellular M.tuberculosis DNA targets bacteria
cdiAMP secreted by intracellular Listeria STING DNA sensing pathway. Science 350, 568571 for autophagy by activating the host DNA-sensing
monocytogenes activates a host typeI interferon (2015). pathway. Cell 150, 803815 (2012).
response. Science 328, 17031705 (2010). 32. Gao,D. etal. Cyclic GMP-AMP synthase is an innate 54. Dey,B. etal. A bacterial cyclic dinucleotide activates
This paper reports that CDNs from bacteria immune sensor of HIV and other retroviruses. the cytosolic surveillance pathway and mediates
activate innate immune signalling. Science 341, 903906 (2013). innate resistance to tuberculosis. Nat. Med. 21,
11. Sun,L., Wu,J., Du,F., Chen,X. & Chen,Z.J. 33. Lahaye,X. etal. The capsids of HIV1 and HIV2 401406 (2015).
Cyclic GMP-AMP synthase is a cytosolic DNA sensor determine immune detection of the viral cDNA by the 55. Archer,K.A., Durack,J. & Portnoy,D.A.
that activates the typeI interferon pathway. innate sensor cGAS in dendritic cells. Immunity 39, STING-dependent typeI IFN production inhibits
Science 339, 786791 (2013). 11321142 (2013). cell-mediated immunity to Listeria monocytogenes.
This study shows that cGAS is a synthase that 34. Herzner,A.M. etal. Sequence-specific activation of PLoS Pathog. 10, e1003861 (2014).
binds dsDNA to generate STING-activating CDNs. the DNA sensor cGAS by Yform DNA structures as 56. Centers for DiseaseControl and Prevention.
12. Diner,E.J. etal. The innate immune DNA sensor found in primary HIV1 cDNA. Nat. Immunol. 16, Prevalence of doctor-diagnosed arthritis and arthritis-
cGAS produces a noncanonical cyclic dinucleotide that 10251033 (2015). attributable activity limitationUnited States,
activates human STING. Cell Rep. 3, 13551361 35. Yoh,S.M. etal. PQBP1 is a proximal sensor of the 20102012.MMWR Morb. Mortal. Wkly Rep. 62,
(2013). cGAS-dependent innate response to HIV1. Cell 161, 869873 (2013).
13. Ablasser,A. etal. cGAS produces a 25linked cyclic 12931305 (2015). 57. Nagata,S. & Kawane,K. Autoinflammation by
dinucleotide second messenger that activates STING. 36. Mankan,A.K. etal. Cytosolic RNA:DNA hybrids endogenous DNA. Adv. Immunol. 110, 139161
Nature 498, 380384 (2013). activate the cGAS-STING axis. EMBO J. 33, (2011).
14. Abe,T. etal. STING recognition of cytoplasmic DNA 29372946 (2014). 58. Janko,C. etal. Inflammatory clearance of apoptotic
instigates cellular defense. Mol. Cell 50, 515 37. Yan,N., Regalado-Magdos,A.D., Stiggelbout,B., remnants in systemic lupus erythematosus (SLE).
(2013). Lee-Kirsch,M.A. & Lieberman,J. The cytosolic Autoimmun. Rev. 8, 912 (2008).
15. Cai,X., Chiu,Y.H. & Chen,Z.J. The cGAS-cGAMP- exonuclease TREX1 inhibits the innate immune 59. Martinez Valle,F., Balada,E., Ordi-Ros,J. &
STING pathway of cytosolic DNA sensing and response to human immunodeficiency virus type1. Vilardell-Tarres,M. DNase 1 and systemic lupus
signaling. Mol. Cell 54, 289296 (2014). Nat. Immunol. 11, 10051013 (2010). erythematosus. Autoimmun Rev. 7, 359363
16. Saitoh,T. etal. Atg9a controls dsDNA-driven 38. Booiman,T., Setiawan,L.C. & Kootstra,N.A. Genetic (2008).
dynamic translocation of STING and the innate variation in Trex1 affects HIV1 disease progression. 60. Rice,G.I., Rodero,M.P. & Crow,Y.J. Human disease
immune response. Proc. Natl Acad. Sci. USA 106, AIDS 28, 25172521 (2014). phenotypes associated with mutations in TREX1.
2084220846 (2009). 39. Bridgeman,A. etal. Viruses transfer the antiviral J.Clin. Immunol. 35, 235243 (2015).
17. Konno,H., Konno,K. & Barber,G.N. Cyclic second messenger cGAMP between cells. Science 61. Gall,A. etal. Autoimmunity initiates in
dinucleotides trigger ULK1 (ATG1) phosphorylation of 349, 12281232 (2015). nonhematopoietic cells and progresses via
STING to prevent sustained innate immune signaling. 40. Gentili,M. etal. Transmission of innate immune lymphocytes in an interferon-dependent autoimmune
Cell 155, 688698 (2013). signaling by packaging of cGAMP in viral particles. disease. Immunity 36, 120131 (2012).
18. Kumar,H., Kawai,T. & Akira,S. Pathogen recognition Science 349, 12321236 (2015). This study shows that STING activation is
by the innate immune system. Int. Rev. Immunol. 30, 41. Zeng,M. etal. MAVS, cGAS, and endogenous responsible for TREX1mediated autoinflamatory
1634 (2011). retroviruses in Tindependent Bcell responses. disease and plausibly AGS.
19. Franchi,L., Munoz-Planillo,R. & Nunez,G. Sensing Science 346, 14861492 (2014). 62. Ahn,J., Ruiz,P. & Barber,G.N. Intrinsic self-DNA
and reacting to microbes through the inflammasomes. 42. Inoue,T. & Tsai,B. How viruses use the endoplasmic triggers inflammatory disease dependent on STING.
Nat. Immunol. 13, 325332 (2012). reticulum for entry, replication, and assembly. J.Immunol. 193, 46344642 (2014).
20. Ahn,J., Gutman,D., Saijo,S. & Barber,G.N. Cold Spring Harb. Perspect. Biol. 5, a013250 This report demonstrates that, in the absence of
STING manifests self DNA-dependent inflammatory (2013). TREX1, self-DNA in macrophages can activate
disease. Proc. Natl Acad. Sci. USA 109, 1938619391 43. Aguirre,S. etal. DENV inhibits typeI IFN production in STING and cause inflammatory diseases, such as
(2012). infected cells by cleaving human STING. PLoS Pathog. AGS.
This report demonstrates that STING activation in 8, e1002934 (2012). 63. Yang,Y.G., Lindahl,T. & Barnes,D.E. Trex1
phagocytes following engulfment of dead cells is 44. Li,X.D. etal. Pivotal roles of cGAS-cGAMP signaling exonuclease degrades ssDNA to prevent chronic
responsible for self-DNA-induced inflammatory in antiviral defense and immune adjuvant effects. checkpoint activation and autoimmune disease.
disease. Science 341, 13901394 (2013). Cell 131, 873886 (2007).

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | DECEMBER 2015 | 769

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

64. Pereira-Lopes,S. etal. The exonuclease Trex1 79. Irrazabal,T., Belcheva,A., Girardin,S.E., Martin,A. & 91. Zitvogel,L., Galluzzi,L., Smyth,M.J. & Kroemer,G.
restrains macrophage proinflammatory activation. Philpott,D.J. The multifaceted role of the intestinal Mechanism of action of conventional and targeted
J.Immunol. 191, 61286135 (2013). microbiota in colon cancer. Mol. Cell 54, 309320 anticancer therapies: reinstating immunosurveillance.
65. Ablasser,A. etal. TREX1 deficiency triggers cell- (2014). Immunity 39, 7488 (2013).
autonomous immunity in a cGAS-dependent manner. 80. Ahn,J., Konno,H. & Barber,G.N. Diverse roles of 92. Hartlova,A. etal. DNA damage primes the type I
J.Immunol. 192, 59935997 (2014). STING-dependent signaling on the development of interferon system via the cytosolic DNA sensor
This study reports that cGAS is involved in cancer. Oncogene 34, 53025308 (2015). STING to promote anti-microbial innate immunity.
STING-dependent autoinflammatory disease 81. Zhu,Q. etal. Cutting edge: STING mediates Immunity 42, 332343 (2015).
that is triggered by TREX1 deficiency. protection against colorectal tumorigenesis by 93. Kondo,T. etal. DNA damage sensor MRE11
66. Gehrke,N. etal. Oxidative damage of DNA confers governing the magnitude of intestinal inflammation. recognizes cytosolic double-stranded DNA and
resistance to cytosolic nuclease TREX1 degradation J.Immunol. 193, 47794782 (2014). induces typeI interferon by regulating STING
and potentiates STING-dependent immune sensing. References 80 and 81 show a protective role for trafficking. Proc. Natl Acad. Sci. USA 110,
Immunity 39, 482495 (2013). STING in recognizing DNA damage and facilitating 29692974 (2013).
67. Kukat,C. & Larsson,N. G. mtDNA makes a Uturn wound repair in the colon. 94. Lemos,H., Huang,L., McGaha,T. & Mellor,A.L.
for the mitochondrial nucleoid. Trends Cell Biol. 23, 82. Gajewski,T.F., Schreiber,H. & Fu,Y.X. Innate and STING, nanoparticles, autoimmune disease and
457463 (2013). adaptive immune cells in the tumor microenvironment. cancer: a novel paradigm for immunotherapy?
68. West,A.P. etal. Mitochondrial DNA stress primes Nat. Immunol. 14, 10141022 (2013). Expert Rev. Clin. Immunol. 11, 155165 (2015).
the antiviral innate immune response. Nature 520, 83. Woo,S.R. etal. STING-dependent cytosolic DNA 95. Zhang,H. etal. Cell-free tumor microparticle vaccines
553557 (2015). sensing mediates innate immune recognition of stimulate dendritic cells via cGAS/STING signaling.
69. White,M.J. etal. Apoptotic caspases suppress immunogenic tumors. Immunity 41, 830842 Cancer Immunol. Res. 3, 196205 (2015).
mtDNA-induced STING-mediated typeI IFN (2014). 96. Dubensky,T.W.Jr., Kanne,D.B. & Leong,M.L.
production. Cell 159, 15491562 (2014). This study shows that antitumour Tcell responses Rationale, progress and development of vaccines
70. Rongvaux,A. etal. Apoptotic caspases prevent the require the activation of STING in phagocytes that utilizing STING-activating cyclic dinucleotide
induction of typeI interferons by mitochondrial DNA. have engulfed tumour cells. adjuvants. Ther. Adv. Vaccines 1, 131143 (2013).
Cell 159, 15631577 (2014). 84. Deng,L. etal. STING-dependent cytosolic DNA 97. Gao,P. etal. Structure-function analysis of STING
References 69 and 70 show that mitochondrial sensing promotes radiation-induced type I activation by c[G(2,5)pA(3,5)p] and targeting by
DNA that has leaked into the cytosol may activate interferon-dependent antitumor immunity in antiviral DMXAA. Cell 154, 748762 (2013).
STING-dependent inflammatory responses. immunogenic tumors. Immunity 41, 843852 98. Jin,L. etal. Identification and characterization of a
71. Creagh,E.M. Caspase crosstalk: integration of (2014). lossoffunction human MPYS variant. Genes Immun.
apoptotic and innate immune signalling pathways. This study shows that radiation-induced tumour 12, 263269 (2011).
Trends Immunol. 35, 631640 (2014). cell death stimulates STING-dependent antitumour 99. Prantner,D. etal. 5,6Dimethylxanthenone4acetic
72. Perl,A., Hanczko,R. & Doherty,E. Assessment of Tcell responses. acid (DMXAA) activates stimulator of interferon gene
mitochondrial dysfunction in lymphocytes of patients 85. Klarquist,J. etal. STING-mediated DNA sensing (STING)-dependent innate immune pathways and is
with systemic lupus erythematosus. Methods Mol. promotes antitumor and autoimmune responses to regulated by mitochondrial membrane potential.
Biol. 900, 6189 (2012). dying cells. J.Immunol. 193, 61246134 (2014). J.Biol. Chem. 287, 3977639788 (2012).
73. Nowarski,R., Gagliani,N., Huber,S. & Flavell,R.A. 86. Roberts,Z.J. etal. The chemotherapeutic agent 100. Liu,Y. etal. Activated STING in a vascular and
Innate immune cells in inflammation and cancer. DMXAA potently and specifically activates the pulmonary syndrome. N.Engl. J.Med. 371, 507518
Cancer Immunol. Res. 1, 7784 (2013). TBK1IRF3 signaling axis. J.Exp. Med. 204, (2014).
74. Salcedo,R., Cataisson,C., Hasan,U., Yuspa,S.H. & 15591569 (2007). This paper reports that mutations in TMEM173
Trinchieri,G. MyD88 and its divergent toll in 87. Corrales,L. etal. Direct activation of STING in the that cause constitutive activity of STING can
carcinogenesis. Trends Immunol. 34, 379389 tumor microenvironment leads to potent and induce inflammatory disease.
(2013). systemic tumor regression and immunity. Cell Rep. 11, 101. Jeremiah,N. etal. Inherited STING-activating
75. Swann,J.B. etal. Demonstration of inflammation- 10181030 (2015). mutation underlies a familial inflammatory syndrome
induced cancer and cancer immunoediting during This study demonstrates that the use of STING with lupus-like manifestations. J.Clin. Invest. 124,
primary tumorigenesis. Proc. Natl Acad. Sci. USA agonists can exert potent antitumour therapeutic 55165520 (2014).
105, 652656 (2008). effects. 102. Lemos,H. etal. Activation of the STING adaptor
76. Ahn,J. etal. Inflammation-driven carcinogenesis is 88. Fu,J. etal. STING agonist formulated cancer attenuates experimental autoimmune encephalitis.
mediated through STING. Nat. Commun. 5, 5166 vaccines can cure established tumors resistant to J.Immunol. 192, 55715578 (2014).
(2014). PD1 blockade. Sci. Transl Med. 7, 283ra52 103. Wu,X. etal. Molecular evolutionary and structural
This study shows that DNA-damaging agents (2015). analysis of the cytosolic DNA sensor cGAS and STING.
can cause DNA leakage in the cytosol to activate 89. Wang,Z. & Celis,E. STING activator cdiGMP Nucleic Acids Res. 42, 82438257 (2014).
STING-mediated inflammatory events and drive enhances the anti-tumor effects of peptide vaccines 104. Mozzi,A. etal. OASes and STING: adaptive evolution
skin cancer. in melanoma-bearing mice. Cancer Immunol. in concert. Genome Biol. Evol. 7, 10161032 (2015).
77. Salcedo,R. etal. MyD88mediated signaling prevents Immunother. 64, 10571066 (2015). 105. Yi,G. etal. Single nucleotide polymorphisms of
development of adenocarcinomas of the colon: role of 90. Ohkuri,T., Ghosh,A., Kosaka,A., Sarkar,S.N. & human STING can affect innate immune response
interleukin 18. J.Exp. Med. 207, 16251636 (2010). Okada,H. Protective role of STING against to cyclic dinucleotides. PLoS ONE 8, e77846 (2013).
78. Huber,S. etal. IL22BP is regulated by the gliomagenesis: rational use of STING agonist in
inflammasome and modulates tumorigenesis in the anti-glioma immunotherapy. Oncoimmunology 4, Competing interests statement
intestine. Nature 491, 259263 (2012). e999523 (2015). The author declares no competing interests.

770 | DECEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved

You might also like