Subido por Ricardo Ponce

Sting in cancer

Anuncio
Oncogene (2015), 1–7
© 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15
www.nature.com/onc
SHORT COMMUNICATION
Diverse roles of STING-dependent signaling on the
development of cancer
J Ahn, H Konno and GN Barber
Stimulator of interferon genes (STING) is a cellular sensor that controls cytosolic DNA-activated innate immune signaling. We have
previously demonstrated that STING-deficient mice are resistant to carcinogen-induced skin cancer, similar to myeloid
differentiation primary response gene 88 (MyD88) deficient mice, since the production of STING-dependent DNA-damage-induced
proinflammatory cytokines, that likely require MyD88 signaling to exert their growth-promoting activity, are prevented. In contrast,
MyD88-deficient mice are sensitive to colitis-associated cancer (CAC), since selected cytokines generated following DNA-damage
also activate repair pathways, which can help prevent tumor development. Here, we demonstrate that STING signaling facilitates
wound repair processes and that analogous to MyD88-deficient mice, STING-deficient mice (SKO) are prone to CAC induced by
DNA-damaging agents. SKO mice harboring tumors exhibited low levels of tumor-suppressive interleukin-22 binding protein
(IL-22BP) compared to normal mice, a cytokine considered critical for preventing colon-related cancer. Our data indicate that STING
constitutes a critical component of the host early response to intestinal damage and is essential for invigorating tissue repair
pathways that may help prevent tumorigenesis.
Oncogene advance online publication, 2 February 2015; doi:10.1038/onc.2014.457
INTRODUCTION
The innate immune system provides the first line of defense
against pathogen infection though can also influence pathways
than can control tumorigenesis.1,2 For example, it is known that
the cellular adapter MyD88 (myeloid differentiation primary
response gene 88) that facilitates Toll-like receptor and
interleukin-1 receptor (IL-1R) signaling pathway in the innate
immune response can regulate tumorigenesis through control of
nuclear factor-κB (NF-κB) activation, cytokine secretion and
inflammation.2 Mice lacking MyD88 are resistant to carcinogen
(7,12-dimethylbenz(a)anthracene (DMBA)) induced skin cancer
since MyD88-controlled cytokine and growth factor production,
which augments skin tumorigenesis is averted.3 Of note is that the
mechanisms underlining carcinogen-driven cytokine production
largely remain unknown.
Stimulator of interferon genes (STING) is a cellular protein that is
critical for activating the production of various cytokines such as
type I interferon in response to the detection of microbial doublestranded DNA (dsDNA) or cyclic dinucleotides (CDNs).4–6 In
addition, self-DNA is also capable of activating STING-dependent
signaling and this pathway has now been shown to play a key role
in the cause of a variety of autoinflammatory diseases.7,8 Recently,
it has been further reported that STING-dependent cytokine
production can be induced by DNA-damaging agents, including
DMBA. It was observed that DMBA caused nucleosome release
into the cytosol to trigger STING activity.9 STING was found to
control the expression of cytokines requiring MyD88 signaling for
the extended production of proinflammatory cytokines such as
tumor necrosis factor α.9 Thus, STING-deficient mice were
observed to be resistant to DMBA-aggravated skin carcinogenesis,
similar to MyD88-deficient mice.9
However, MyD88-deficient mice are known to be susceptible to
colitis-associated carcinogenesis (CAC) induced by drugs such as
azoxymethane (AOM) and dextran sulfate sodium (DSS).10 AOM is
the metabolite of 1,2-dimethylhydrazine (DMH) and is converted
to methylazoxymethanol, which mediates O-methylguanine formation to trigger DNA damage responses.11 A single injection of
AOM into mice, followed by administration of the inflammatory
agent DSS via drinking water induces almost 100% colon cancer.
In this situation, MyD88 exerts a protective effect in part by
facilitating the production of interleukin-18 (IL-18), in epithelial
cells, which downregulates dendritic cell production of the
interleukin-22 (IL-22) binding protein (IL-22BP).12 IL-22BP suppresses the function of IL-22 which is produced from innate
lymphoid cells in response to cellular/tissue damage and which
potently stimulates the proliferation of intestinal epithelial cells.
We have previously demonstrated that the cellular protein
STING facilitates cytosolic DNA-triggered innate immune signaling
pathways, independent of Toll-like receptor 9 or the DNA sensor
absent in melanoma 2 (AIM II).13 In humans, STING is a 348 amino
acid endoplasmic reticulum-associated molecule predominantly
expressed in epithelial cells as well as cells of the hematopoietic
lineage, that has been shown to play a key role in triggering
innate immune signaling pathways in response to infection by
viruses such as herpes simplex virus 1, and even bacteria.4 STING
has also been shown to be responsible for triggering vascular and
pulmonary syndrome, self-DNA-induced inflammatory diseases
such as Aicardi–Goutieres syndrome and perhaps forms of severe
systemic lupus erythematosus.8,14–16 STING may associate
with dsDNA species directly and is highly activated by CDNs
(cyclic di-GMP-AMP[cGAMP-c(G(2′,5′)pA(3′,5′)p)]) generated by
certain bacteria or by cytosolic dsDNA triggering the activation
Department of Cell Biology and Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Miami, FL, USA. Correspondence: Dr GN Barber, Department of
Cell Biology and Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, 511 Papanicolaou Building, 1550 NW 10th Ave, Miami, FL 33136, USA.
E-mail: [email protected]
Received 30 October 2014; revised 18 November 2014; accepted 26 November 2014
Role of STING-dependent signaling on cancer
J Ahn et al
2
of a synthase, referred to as cGAS (cyclic GMP-AMP synthase,
C6orf150, Mab-21 domain-containing protein).6
Given that STING appears to play a pivotal role in controlling a
variety of inflammation-driven events and may control MyD88-
dependent carcinogen-induced skin cancer, we wished to
address the role of STING in inflammation-aggravated intestinal
tumorigenesis. Using the AOM/DSS model, we observed that
similar to MyD88, STING-deficient mice (SKO) are sensitive to CAC
Mo
H
DM
ck
Mo
H
DM
Rgs1
Ccl3
Cxcl2
EG546166
Rgs1
Atf3
Rgs1
Gdf15
Ccl4
EG434760
Aifm2
lgtp
Sprr2a
Rab10
Scyl1
Zfp566
Mcm7
4930476L21Rik
Sec11c
Usp18
Ube2t
Cxcl2
Klhdc4
Slfn2
Pdhb
Serhl
LOC280205
Rdh1
Cyp11b1
Frag1
Josd3
Nkain2
1110059G10Rik
1810034K20Rik
Afg312
Coro1b
Rnf121
Snx16
Cyr61
Gbp3
Ngfb
LOC100038882
Myocd
Cdkn1a
Bet1
Asb16
Ppm1f
2510040D07Rik
Tapbpl
Rps6kb1
Cxcl10 fold changes
Cxcl10 fold changes
8
6
4
2
0
AOM
1
0
WT
SKO
5
Mock
AOM
4
3
2
1
0
10
WT
SKO
*
Mock
DMH
8
6
4
2
0
2
WT
SKO
*
Mock
DMH
1.5
1
0.5
WT
SKO
DMH
3
siNS
siSting
*
2
1
0
*
1.2
1.0
0.8
0.6
0.4
0.2
0.0
Mock
AOM
SKO colon
DMH
siNS
siSting
Human normal colon
Colon
WT colon
2
FHC
*
Mock
Mock
AOM
3
0
FHC
10
*
4
1.04
0.69
0.35
0.00
0.35
0.69
1.04
1.04
0.69
0.35
0.00
0.35
0.69
1.04
EG546166
Ifit3
Usp18
Rnf2
LOC667370
Ifit3
Usp18
LOC100048346
9930013L23Rik
Gm166
9530096D07Rik
C530043K16Rik
LOC674960
LOC100038882
V1rh12
5730455P16Rik
Chst5
Fn1
9630025C19Rik
A530082L16Rik
S100a16
Ndufb6
Cd83
4732474K08Rik
Calm2
AU019823
9130230L23Rik
2200002D01Rik
Ogfod2
Rnf213
Rsad2
Igf2bp1
Gm50
LOC100039693
Peg13
Olfr67
Gm1381
Igtp
Myo18b
2700078K21Rik
2900097C17Rik
Zc3h10
Eif2c3
Ensa
Ell2
Efna5
Chrna2
Kctd12b
Catnb
2410002F23Rik
5
Cxcl10 fold changes
ck
ck OM
M
AO Mo
A
SKO
Ifit3 fold changes
WT
Cxcl10 fold changes
ck
Mo
SKO
Sting fold changes
WT
Ifit3 fold changes
MEF
Oncogene (2015), 1 – 7
© 2015 Macmillan Publishers Limited
Role of STING-dependent signaling on cancer
J Ahn et al
3
indicating a protective role for STING in tumorigenesis. Our data
indicate that STING may be a key sensor that promotes the
elimination of damaged intestinal epithelial cells.
RESULTS AND DISCUSSION
Activation of STING-dependent genes by AOM
Given that chronic inflammation is known to aggravate colon
cancer and that STING has been shown to influence inflammatory
responses, especially those invoked by cytosolic self or pathogenrelated DNA, we examined the role of STING in the control of
inflammatory CAC.1,2,15 Toward these objectives, we utilized the
AOM/DSS model and analyzed the effects of AOM and precursor
DMH on STING signaling. Principally, wild-type (WT) or STINGdeficient (SKO) murine embryonic fibroblasts (MEFs) were treated
in vitro with DMH or metabolite AOM for 8 h, and microarray
analysis performed to analyze the consequences to gene
expression. This study indicated that AOM activated mRNA
production of a wide array of innate immune-related genes in
WT cells, including interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) and chemokine (C-X-C motif) ligand 2
(CXCL2) (Figure 1a and Supplementary Figure 1). However, there
was a marked decrease in the production of the same genes in
SKO MEFs indicating that AOM was indeed capable of activating
the STING pathway (Figure 1a, left panel). A similar effect was
observed following the treatment of cells with DMH (Figure 1a,
right panel). STING-dependent gene expression was confirmed
following RT–PCR analysis of mRNA representing Cxcl10 and IFIT3
(Figure 1b). We similarly treated normal human colon epithelial
cells (FHCs) with AOM and observed a comparable induction of
innate immune genes, controlled by STING, including Cxcl10
(Figure 1c). The production of Cxcl10 by AOM was similarly
reduced in FHCs treated with RNAi to STING (Figure 1d). To
determine the consequences of DMH/AOM treatment on STING’s
ability to activate these key transcription factors, we carried out
immunofluorescence analysis on MEFs and FHCs treated with
these drugs. This study indicated that DMH/AOM could instigate
the translocation of interferon regulatory factor 3/NF-κB in treated
cells (Supplementary Figure 2). Thus, the DNA-damaging agent
DMH/AOM can invoke STING-dependent signaling.
Loss of STING renders mice susceptible to CAC
Our data indicate that DMH/AOM can activate STING in vitro. To
examine the consequences of this in vivo, we treated mice once
with AOM and subsequently orally with four treatments of DSS.
Prior to this, we analyzed STING expression in the intestine by
immunohistochemistry analysis. This study showed that STING
was expressed in lamina propria cells as well as in endothelial and
epithelial cells of the gastrointestinal tract (Figure 1e). After
13 weeks the mice were analyzed for tumor development in the
colon. Surprisingly, we observed that SKO mice developed colonic
tumors at a much higher frequency compared to WT mice
(Figures 2a–c and Supplementary Figure S3). Indeed, 4/7 WT mice
exhibited tumor formation compared to 7/7 SKO within the same
time period (Figures 2b and c and Supplementary Figure S3a).
Hematoxylin and eosin analysis confirmed that AOM/DSS-treated
SKO mice exhibited significant inflammatory cell infiltration and
development of adenocarcinoma in the colon, compared to
similarly treated WT mice (Figures 2d and e and Supplementary
Figure S3b). However, Microarray analysis indicated that tumors
from WT mice exhibited higher levels of selected gene expression,
such as Cxcl13 and Ccr6, compared to tumors retrieved from SKO
mice, perhaps since loss of STING suppressed immunomodulatory
transcriptional events (Figures 2f–h). Collectively, we postulate
that STING may recognize damaged DNA and activate the
production of cytokines that conceivably could promote tissue
repair or stimulate the immune system to eradicate such cells.
Thus, loss of STING may enable damaged cells to escape immune
surveillance processes and progress more readily into tumors.
Suppression of IL-22BP expression in STING-deficient mice
Although, we demonstrated that loss of STING facilitated colon cancer
development, the tumor-suppressive mechanisms associated with
STING activity remain to be clarified. It remained plausible that STING
could exert direct tumor-suppressive, growth inhibitory or proapoptotic properties similar to tumor suppressor p53. Further, AOM
treatment has been known to induce frequent Ras mutations, which in
the context of loss of STING, could facilitate cellular transformation.17
Expression of oncogenic Ras in an environment where p53 function is
lost renders normal murine cells, the ability to form foci in soft agar
and to become tumorigenic in vivo.18 To evaluate the anti-oncogenic
role of STING further, we transfected WT, SKO or p53-deficient MEFs,
as positive controls, with Myc or activated Ras and monitored cellular
transformation. MEFs lacking p53 were found to be readily
transformed by the introduction of Myc or activated Ras
(Supplementary Figure S4). However, MEFs lacking STING did not
appear appreciably sensitive to transformation following overexpression of Myc or activated Ras. Thus, the absence of STING does not
appear to exert an oncogenic stimulus, at least in vitro or to cooperate
with Myc or Ras in the cellular transformation process.
However, it has been demonstrated that mice lacking certain
cytokines such as IL-18, IL-22 or the innate immune adapter
MyD88 are similarly susceptible to AOM/DSS-induced CAC.1,2,10,12
In this situation, MyD88 exerts a protective effect by facilitating
the production of IL-18 through the IL-18R, which is required to
inhibit IL-22BP.3,12,19–22 IL-22BP is necessary to suppress IL-22
function, which can promote the proliferation of intestinal
epithelial cells following damage by carcinogens or inflammatory
agents.12 Mice lacking IL-18 or IL-22BP are highly susceptible to
CAC, similar to STING-deficient mice.12 To examine whether AOM/
DSS treatment could affect IL-18 and IL-22BP expression in SKO
mice, we analyzed IL-18 and IL-22BP levels in untreated normal
mice (control) or tumor tissue taken from the AOM/DSS-treated
WT or SKO mice. We observed that IL-18 expression was slightly
upregulated in tumor tissue retrieved from WT mice treated with
AOM/DSS (Figure 2i). We observed that IL-22BP expression levels
Figure 1. Activation of STING-dependent genes by AOM. (a) Gene array analysis of wild-type (WT) and STING-deficient (SKO) mouse
embryonic fibroblasts (MEFs) treated with AOM at 0.14 mM for 8 h (left) and DMH at 1 mM for 8 h (right). MEFs were obtained from embryos
from WT and SKO mice at embryonic 15 days by a standard procedure as described (Ishikawa 2008). Gene array analysis was examined by
Illumina Sentrix BeadChip array (Mouse WG6 version 2). Highest variable genes are shown. Rows represent individual genes and columns
represent individual samples. Pseudo-colors indicate transcript levels below (green), equal to (black) or above (red) the mean. Scale represents
the intensity of gene expression (log2 scale ranges between − 2.4 and 2.4). (b) Quantitative real-time PCR analysis (qPCR) of Cxcl10 and Ifit3 in
MEFs treated with AOM and DMH same as a. Total RNA was reverse transcribed using moloney murine leukemia virus
(M-MLV) reverse transcriptase. qPCR was performed using TaqMan gene expression assay (Cxcl10: Mm00445235, Ifit3: Mm0170846).
(c) qPCR analysis of Cxcl10 in human epithelial cell (FHC) treated with AOM and DMH at 1 mM for 24 h. (d) FHCs were transfected with STING or
control siRNA for 72 h followed by AOM and DMH treatment same as c, and were then subjected to Cxcl10 mRNA expression (left). STING
expression level after siRNA treatment was determined by qPCR (right). Data are representative of at least two independent experiments. Error
bars indicate s.d. *P o0.05, Student’s t-test. (e) Immunohistochemistry staining of the colon tissue from WT and SKO mice (left) and human
(right). All images were shown at original magnification, x200.
© 2015 Macmillan Publishers Limited
Oncogene (2015), 1 – 7
Role of STING-dependent signaling on cancer
J Ahn et al
4
W1
DSS
DSS
W4
DSS
W7
W10
AOM
injection
WT Cont
SKO Cont
160
Body weight (%)
DSS
W13
Tumor
evaluation
WT AOM/DSS
SKO AOM/DSS
140
120
100
80
0
4
8
23
28
36
45
49
62
72
77
106
121
60
SKO
WT
6
DSS/AOM
DW
DSS/AOM
Number of
polyps
DW
Microendoscopy
Cont
DSS/AOM
4
2
Inflammation score
0
HE
4
Control
DSS/AOM
3
2
1
0
WT
OM
l
n
tro
Co
A
S/
DS
Co
Symbol
OM
l
ro
nt
DS
3.00
2.00
1.00
0.00
1.00
2.00
3.00
*
80
Control
AOM/DSS
60
40
20
SKO
5
IL18 Fold changes
100
4
3
WT
SKO
Control
*
AOM/DSS
*
2
1
WT
SKO-DW
148.69
58.01
28.63
28.13
24.57
17.66
16.98
16.82
16.59
15.42
14.57
14.06
13.72
13.20
13.03
12.86
12.44
12.37
11.53
11.17
11.16
11.13
10.91
10.88
10.86
10.75
10.44
10.41
10.16
10.15
10.11
9.54
9.51
9.50
9.48
9.42
9.21
9.19
9.19
9.17
9.10
9.06
8.95
8.82
8.42
8.39
8.08
7.93
7.91
7.89
7.86
0
0
WT
Oncogene (2015), 1 – 7
Control
AOM/DSS
CCR6 Fold changes
Cxcl13 Fold changes
*
WT-DSS/AOM
1810030J14Rik
1810030J14Rik
Atp12a
LOC673501
Faim3
LOC100047788
Cd79b
Fcrla
Ighg
Insl5
Cxcl13
AI324046
H2-Ab1
Dlk1
LOC100046120
LOC100047815
Slpi
Cd52
Cd52
Cd74
AI854703
Cd79b
Ela1
D930028F11Rik
Klhl6
Klhl6
Cd74
H2-Oa
Lyz2
Ttr
Fcrla
B3gnt7
Adh1
Tnfrsf13c
B3gnt5
Ctse
Coro1a
AI324046
Igh-6
Fcrla
Ccr6
Dok3
H2-Ob
Mfge8
Igh-6
Arhgdib
Coro1a
H2-DMb2
Hvcn1
Nt5e
Mfge8
A
S/
1810030J14Rik
1810030J14Rik
Atp12a
LOC673501
Faim3
LOC100047788
Cd79b
Fcrla
lghg
lnsl5
Cxcl13
Al324046
H2-Ab1
Dlk1
LOC100046120
LOC100047815
Slpi
Cd52
Cd52
Cd74
Al854703
Cd79b
Ela1
D930028F11Rik
Klhl6
Klhl6
Cd74
H2-Oa
Lyz2
Ttr
Fcrla
B3gnt7
Adh1
Tnfrsf13c
B3gnt5
Ctse
Coro1a
Al324046
lgh-6
Fcrla
Ccr6
Dok3
H2-Ob
Mfge8
lgh-6
Arhgdib
Coro1a
H2-DMb2
Hvcn1
Nt5e
Mfae8
18
16
14
12
10
8
6
4
2
0
SKO
SKO
SKO
IL22bp Fold changes
WT
8
7
6
5
4
3
2
1
0
31.43
12.10
9.28
5.32
1.19
-1.19
1.09
1.04
-1.30
3.84
1.14
1.06
1.01
1.92
1.77
1.36
1.64
-1.06
-1.41
-1.19
5.50
-1.00
4.65
-1.44
-1.05
-1.04
-1.16
1.11
-1.25
3.68
-1.06
3.77
2.01
1.05
2.88
3.35
-1.13
-1.09
-1.22
1.05
1.14
1.19
1.11
1.16
-1.29
-1.22
-1.35
-1.12
1.12
3.48
1.11
*
WT
SKODSS/AOM
5.58
3.17
18.66
5.11
2.42
1.09
1.56
1.64
1.31
1.79
1.40
1.00
5.73
1.08
10.48
1.59
18.37
3.56
2.68
6.07
4.12
1.42
4.55
1.16
2.16
1.71
5.37
1.85
6.48
3.02
1.35
5.13
1.41
1.39
10.19
10.23
2.84
-1.08
1.51
1.42
1.79
2.40
1.23
4.50
1.38
2.88
2.70
2.99
1.95
12.67
4.28
Control
AOM/DSS
SKO
© 2015 Macmillan Publishers Limited
Role of STING-dependent signaling on cancer
J Ahn et al
5
were also elevated in the tumors of AOM/DSS-treated WT mice.
In contrast, we noted the significantly reduced expression of IL-18
and IL-22BP in the tumors of SKO mice, when compared to WT
mice (Figure 2i). The expression of IL-22 was seen to remain
relatively unaffected (data not shown). It was surprising to note
that IL-22BP levels were elevated in the presence of IL-18.
However, it has been reported that the downregulation of IL-22BP
can occur even in the absence of IL-18, indicating that alternate
STING-induced cytokines may clearly contribute to the regulation
of IL-22BP.12
In addition, we noted from our microarray analyses that IL-18
levels were reduced in SKO MEFs treated with STING-activating
dsDNA (dsDNA90 base pairs; Figure 3a, left panel). We therefore
investigated the involvement of STING on the possible regulation
of the IL-18/IL-22BP axis. First, we confirmed the influence of
STING on IL-18 expression, since we additionally noted that the
promoter of this cytokine is known to harbor numerous sites
recognized by innate immune gene-activating transcription
factors such as signal transducer and activator of transcription 1,
NF-κB, IRF1 and IRF7 (Supplementary Figure S5). Our analysis
indicated that IL-18, which is expressed in a wide variety of cell
types, is a STING inducible gene, as determined following
treatment of MEF cells and bone marrow-derived dendritic cells
(BMDCs) with dsDNA or STING-activating CDNs (cGAMP; Figure 3a,
middle and right panel). A similar study indicated that DMH/AOM
could also trigger the production of IL-18 in dendritic cells, in a
STING-dependent manner (Figure 3b). To further this study, we
examined IL-22BP expression in BMDCs stimulated with IL-18
MEF
BMDCs
Gene array
RT PCR
WT
SKO
3
2
1
0
t
0
on
A9
C
N
8
7
6
5
4
3
2
1
0
*
k
oc
0
A9
M
D
ds
WT
SKO
N
lF
RT PCR
*
ds
D
N
7
6
5
4
3
2
1
0
lL18 fold changes
*
lL18 Fold changes
lL18 Fold changes
4
0
A9
M
cG
N
D
ds
P
AM
cG
BMDCs
6
WT
SKO
*
lL22bp Fold changes
lL18 Fold changes
*
k
oc
P
AM
BMDCs
16
14
12
10
8
6
4
2
0
*
WT
SKO
5
Mock
lL18
*
4
3
2
1
0
No
AOM
DMH
WT
SKO
Figure 3. IL-18 controls IL-22BP in STING-dependent manner. (a) Fold changes from gene array analysis of IL-18 in WT and SKO MEFs
administered with 4 ug/ml of dsDNA90 and IFNβ for 8 h (left). qPCR analysis of IL18 in WT and SKO MEFs, and bone marrow-derived dendritic
cells (BMDCs) transfected with 4 μg/ml of dsDNA90 and cyclic-di-GMP-AMP (cGAMP) for 8 h (middle). (b) qPCR analysis of IL-18 in WT and SKO
BMDCs with 1 mM of AOM and 1 mM of DMH for 8 h. (c) qPCR analysis of IL-22BP in WT and SKO BMDCs treated with 10 ng/ml of recombinant
mouse IL-18 protein (MBL B002-5) for 24 h. Total RNA was reverse transcribed using M-MLV reverse transcriptase. qPCR was performed using
TaqMan gene expression assay (IL-18: Mm00434225, IL-22BP: Mm01192969). Data are the mean of at least five mice. Error bars indicate s.d.
*P o0.05, Student’s t-test.
Figure 2. Loss of STING renders mice susceptible to CAC. (a) Schematic representation and body weight of AOM/DSS-induced colitis model.
WT (n = 7) and SKO (n = 7) mice (B6;129 mix) were intravenously injected with AOM at a dose 10 mg/kg. After 1 day, mice were fed 5% DSS in
drinking water for 7 days. This cycle was repeated four times. Normal drinking water was used for control group. At 91 days, micro-endoscopic
procedure was performed in a blinded manner for counting number of polyps. Mice were killed at day 121 and the colon was resected,
flushed with phosphate-buffered saline, fixed in formalin for histology or froze for RNA expression analysis. Representative photographs of
macro-endoscopic colon tumors (b) and number of polyps (c). Hematoxylin and eosin staining (d) of WT (n = 7) and SKO (n = 7) mice either
AOM/DSS treated or normal water treated and inflammation score (0: normal to 3: most severe; e). (f) Gene array analysis of normal tissue
(control) and tumor tissue (DSS/AOM) from WT and SKO mice treated same as a. Rows represent individual genes and columns represent
individual samples. Pseudo-colors indicate transcript levels below (green), equal to (black) or above (red) the mean. Scale represents the
intensity of gene expression (log2 scale ranges between − 2.4 and 2.4). (g) Highest variable gene lists are shown. (h) qPCR of Cxcl13 and Ccr6
in normal tissue (control) and tumor tissue (DSS/AOM) from WT and SKO mice treated same as a. (i) qPCR analysis of IL-18 and IL-22BP (IL-18:
Mm00434225, IL-22BP: Mm01192969) from normal tissue (control) and tumor tissue (DSS/AOM) from WT and SKO mice. Data are the mean of
at least five mice. Error bars indicate s.d. *P o0.05, Student’s t-test.
© 2015 Macmillan Publishers Limited
Oncogene (2015), 1 – 7
Role of STING-dependent signaling on cancer
J Ahn et al
6
recombinant protein and observed the upregulation of IL-22BP
in WT BMDCs compared to SKO BMDCs (Figure 3c and
Supplementary Figure S6). These data are similar to Figure 2i
showing that IL-18 and IL-22BP expression levels were also lower
in the tumors of AOM/DSS-treated SKO mice compared to the
tumors of WT mice (Figure 2i). These data suggest that STING may
regulate IL-22BP expression by IL-18.
Taken together, it is conceivable that DNA damage or the
sensing of microbial ligands that may invade damaged colon
tissue can trigger STING activity leading to the activation of
inflammatory wound repair initiating cytokines as well as the
indirect suppression of growth inhibitory IL-22BP. Our data thus
indicate that similar to mice lacking MyD88, IL-18 or IL-22BP,
STING-deficient mice are also prone to CAC induced by AOM/DSS.
Although, we demonstrate here a protective role for STING in
the prevention of CAC induced by AOM/DSS carcinogenic
treatment, these findings are in contrast to our previous data
indicating that STING-deficient mice are resistant to DMBAinduced skin cancer.9 In this scenario, DNA-leaked from the
nucleus of carcinogen damaged dermal-associated cells activates
STING-dependent cytokine production, intrinsically, to attract
infiltrating phagocytes. Such phagocytes engulf dying cells to
activate extrinsic STING-dependent cytokine production, which
further drives inflammatory processes that promote tumor
development.9 This situation is reminiscent of similarly treated
MyD88-deficient mice, which are also resistant to DMBAaggravated skin cancer. Our data indicate that STING triggers
the production of cytokines that bind to receptors requiring
MyD88-dependent signaling for additional cytokine production,
such as tumor necrosis factor α.3 Our data here conversely indicate
that STING-driven cytokine production can protect against the
development of other types of tumors, such as carcinogeninfluenced colon cancer.23 This event may occur in part through
STING's ability to control the production of IL-22BP, although
STING clearly controls the production of numerous other
regulatory proteins and cytokines. Following tissue damage, for
example by DSS, it has been reported that IL-22 is induced and
manifests protective, wound-healing effects, including the promotion of tissue regeneration.1,2,10,12 However, if left uncontrolled,
IL-22 can also endorse tumor development.12 IL-22 is therefore
tightly regulated by secreted IL-22BP, which is expressed by
CD11c+ dendritic cells.12 The importance of IL-22BP in controlling
IL-22 has been emphasized through observing that IL-22BPdeficient mice are also susceptible to AOM/DSS-induced CAC,
similar to STING-deficient mice.12 Nevertheless, IL-22 may have
dual functions since mice lacking IL-22 have also been reported to
exhibit enhanced inflammatory responses when treated repeatedly with DSS, plausibly because complete loss of IL-22 may cause
a delay in intestinal repair, which in turn may actually aggravate
inflammatory processes.1,2,10,12 The production of IL-22BP can be
suppressed by IL-18, which is known to be induced early after
DSS-induced intestinal damage. Accordingly, IL-18-deficient mice
are also susceptible to colon cancer, presumably through chronic
suppression of IL-22 activity, by unregulated IL-22BP, which may
mimic the situation observed with IL-22-deficient mice.12 Nevertheless, the control of IL-22BP remains to be fully clarified since
downregulation of IL-22BP has also been reported to occur in the
absence of IL-18.12 In addition, it is known that loss of the Toll-like
receptor and IL-1R/IL-18R adapter MyD88 also renders mice
sensitive to CAC, in part due to loss of IL-18R signaling.20,21 Finally,
susceptibility to AOM/DSS-induced CAC has been shown to be
enhanced in mice lacking caspase-1, the adapter PYCARD
(apoptosis-associated speck-like protein containing a caspase
activation and recruitment domain; ASC) or nucleotide-binding
domain, leucine rich repeat and pyrin domain-containing proteins
3 and 6 (NLRP3/6), presumably since Pro-IL-18 produced by
epithelial cells or dendritic cells requires cleavage prior to
secretion into an active form.24–26
Oncogene (2015), 1 – 7
Our data here indicate that IL-18 and a variety of other
cytokines and chemokines are inducible by dsDNA or CDN’s, or by
AOM/DMH in a STING-dependent manner. Similar to the situation
with IL-22, we propose that intestinal damage triggers STING
activity (as a consequence of DNA damage or even from microbial
ligands such as secreted CDNs or genomic DNA). This results in the
downregulation of IL-22BP, which would enable IL-22 to promote
tissue repair. However, similar to the situation with IL-22, longterm loss of STING may delay wound repair, facilitate microbial
invasion and chronic inflammation, which would actually aggravate tumorigenesis.1,2,12 In addition, loss of STING may enable
damaged cells to escape antitumor immunosurveillance.2 It was
noted that IL-18 expression was not totally ablated in tumors from
SKO mice, presumably since the expression of this cytokine could
be induced by other pathways.12 Despite this, IL-22BP levels
remained low in SKO mice evidently demonstrating the importance of STING in IL-22BP regulation. Collectively, our data indicate
that STING plays a key role in controlling intestinal tissue damage
and CAC in part through regulating IL-22BP’s suppression of
IL-22. Our data also provide a mechanistic explanation for
carcinogenic-induced immunomodulatory effects that can
influence tumorigenesis.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
ACKNOWLEDGEMENTS
We thank Dr Masayuki Fukata for micro-endoscopy analysis; Ms Delia Gutman and Ms
Auristela Rivera for mice breeding; Dr Tianli Xia for helping with confocal analysis; Dr
Biju Issac of the Sylvester Comprehensive Cancer Center Bioinformatics Core Facility
for Gene expression array analysis; Dr Phillp Ruiz and Ms Dayami Hernandez for
helping with immunohistochemistry.
REFERENCES
1 Goldszmid RS, Trinchieri G. The price of immunity. Nat Immunol 2012; 13:
932–938.
2 Nowarski R, Gagliani N, Huber S, Flavell RA. Innate immune cells in inflammation
and cancer. Cancer Immunol Res 2013; 1: 77–84.
3 Cataisson C, Salcedo R, Hakim S, Moffitt BA, Wright L, Yi M et al. IL-1R-MyD88
signaling in keratinocyte transformation and carcinogenesis. J Exp Med 2012; 209:
1689–1702.
4 Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNA-mediated, type I
interferon-dependent innate immunity. Nature 2009; 461: 788–792.
5 Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B, Hyodo M et al. STING is
a direct innate immune sensor of cyclic di-GMP. Nature 2011; 478: 515–518.
6 Cai X, Chiu YH, Chen ZJ. The cGAS-cGAMP-STING pathway of cytosolic DNA
sensing and signaling. Mol Cell 2014; 54: 289–296.
7 Ahn J, Ruiz P, Barber GN. Intrinsic self-DNA triggers inflammatory disease
dependent on STING. J Immunol 2014; 193: 4634–4642.
8 Gall A, Treuting P, Elkon KB, Loo YM, Gale M Jr, Barber GN et al. Autoimmunity
initiates in nonhematopoietic cells and progresses via lymphocytes in an
interferon-dependent autoimmune disease. Immunity 2012; 36: 120–131.
9 Ahn J, Xia T, Konno H, Konno K, Ruiz P, Barber GN. Inflammation-driven carcinogenesis is mediated through STING. Nat Commun 2014; 5: 5166.
10 Salcedo R, Cataisson C, Hasan U, Yuspa SH, Trinchieri G. MyD88 and its divergent
Toll in carcinogenesis. Trends Immunol 2013; 34: 379–389.
11 De Robertis M, Massi E, Poeta ML, Carotti S, Morini S, Cecchetelli L et al. The AOM/
DSS murine model for the study of colon carcinogenesis: from pathways to
diagnosis and therapy studies. J Carcinog 2011; 10: 9.
12 Huber S, Gagliani N, Zenewicz LA, Huber FJ, Bosurgi L, Hu B et al. IL-22BP is
regulated by the inflammasome and modulates tumorigenesis in the intestine.
Nature 2012; 491: 259–263.
13 Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates
innate immune signalling. Nature 2008; 455: 674–678.
14 Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, Montealegre Sanchez GA et al.
Activated STING in a vascular and pulmonary syndrome. N Engl J Med 2014; 371:
507–518.
15 Ahn J, Gutman D, Saijo S, Barber GN. STING manifests self DNA-dependent
inflammatory disease. Proc Natl Acad Sci USA 2012; 109: 19386–19391.
© 2015 Macmillan Publishers Limited
Role of STING-dependent signaling on cancer
J Ahn et al
7
16 Namjou B, Kothari PH, Kelly JA, Glenn SB, Ojwang JO, Adler A et al. Evaluation of
the TREX1 gene in a large multi-ancestral lupus cohort. Genes Immun 2011; 12:
270–279.
17 Jacoby RF, Llor X, Teng BB, Davidson NO, Brasitus TA. Mutations in the K-ras
oncogene induced by 1,2-dimethylhydrazine in preneoplastic and neoplastic rat
colonic mucosa. J Clin Invest 1991; 87: 624–630.
18 Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes
premature cell senescence associated with accumulation of p53 and p16INK4a.
Cell 1997; 88: 593–602.
19 Swann JB, Vesely MD, Silva A, Sharkey J, Akira S, Schreiber RD et al.
Demonstration of inflammation-induced cancer and cancer immunoediting during primary tumorigenesis. Proc Natl Acad Sci USA 2008; 105:
652–656.
20 Salcedo R, Worschech A, Cardone M, Jones Y, Gyulai Z, Dai RM et al.
MyD88-mediated signaling prevents development of adenocarcinomas of the
colon: role of interleukin 18. J Exp Med 2010; 207: 1625–1636.
21 Zenewicz LA, Yancopoulos GD, Valenzuela DM, Murphy AJ, Stevens S, Flavell RA.
Innate and adaptive interleukin-22 protects mice from inflammatory bowel
disease. Immunity 2008; 29: 947–957.
22 Rakoff-Nahoum S, Medzhitov R. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science 2007; 317: 124–127.
23 Zhu Q, Man SM, Gurung P, Liu Z, Vogel P, Lamkanfi M et al. Cutting edge:
STING mediates protection against colorectal tumorigenesis by governing the
magnitude of intestinal inflammation. J Immunol 2014; 193: 4779–4782.
24 Fukata M, Vamadevan AS, Abreu MT. Toll-like receptors (TLRs) and Nod-like
receptors (NLRs) in inflammatory disorders. Semin Immunol 2009; 21: 242–253.
25 Allen IC, TeKippe EM, Woodford RM, Uronis JM, Holl EK, Rogers AB et al.
The NLRP3 inflammasome functions as a negative regulator of tumorigenesis
during colitis-associated cancer. J Exp Med 2010; 207: 1045–1056.
26 Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ et al. NLRP6
inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011;
145: 745–757.
Supplementary Information accompanies this paper on the Oncogene website (http://www.nature.com/onc)
© 2015 Macmillan Publishers Limited
Oncogene (2015), 1 – 7
Descargar