ArticlePDF Available

Oncogenic Ras induces inflammatory cytokine production by up-regulating the squamous cell carcinoma antigens SerpinB3/B4

Authors:

Abstract and Figures

Mounting evidence indicates that oncogenic Ras can modulate cell autonomous inflammatory cytokine production, although the underlying mechanism remains unclear. Here we show that squamous cell carcinoma antigens 1 and 2 (SCCA1/2), members of the Serpin family of serine/cysteine protease inhibitors, are transcriptionally upregulated by oncogenic Ras via MAPK and the ETS family transcription factor PEA3. Increased SCCA expression leads to inhibition of protein turnover, unfolded protein response, activation of NF-κB and is essential for Ras-mediated cytokine production and tumour growth. Analysis of human colorectal and pancreatic tumour samples reveals a positive correlation between Ras mutation, enhanced SCCA expression and IL-6 expression. These results indicate that SCCA is a Ras-responsive factor that plays an important role in Ras-associated cytokine production and tumorigenesis.
SCCA promotes cytokine production by inducing UPR. (a,b) Vector-control or HRasV12-expressing IMR90 cells were stably infected with lentiviral shRNA control (shNTC) or two independent hairpins targeting SCCA. Whole-cell lysates were analysed by western blot with indicated antibodies. Quantification of the ubiquitin blot in a was performed by Image J using the full-length of each lane. (c–g) Vector-control or HRasV12 IMR90 cells were stably infected with lentiviral shRNA control (shNTC) or shRNA hairpins targeting ATF6 or XBP1. (c,g) Whole-cell lysates were analysed by western blot with indicated antibodies. (d) Culture media were collected and subjected to a cytokine antibody array. The blots of indicated cytokines are shown. (e) The relative amount of cytokines was quantified and normalized to that of RasV12-shNTC cells. (f) Total RNA was extracted and cytokine transcript levels were analysed via qRT–PCR and normalized against that in RasV12-shNTC cells. Data shown are mean+s.e.m. of three independent experiments performed in triplicate. (h,i) Vector-control or HRasV12 IMR90 cells were stably infected with lentiviral shRNA control (shNTC) or shSCCA, together with vector-control or XBP1s-expressing construct. (h) Whole-cell lysates were analysed by western blot with indicated antibodies. (i) Total RNA was extracted and cytokine transcript levels were analysed via qRT–PCR and normalized against that in RasV12-shNTC cells. Data shown are mean+s.e.m. of three independent experiments performed in triplicate. **P<0.01; ***P<0.001; NS, non-significant by t-test.
… 
Content may be subject to copyright.
ARTICLE
Received 29 Oct 2013 |Accepted 26 Mar 2014 |Published 23 Apr 2014
Oncogenic Ras induces inflammatory cytokine
production by upregulating the squamous cell
carcinoma antigens SerpinB3/B4
Joseph M. Catanzaro1,*, Namratha Sheshadri1,*, Ji-An Pan1, Yu Sun1, Chanjuan Shi2, Jinyu Li3, R. Scott Powers3,
Howard C. Crawford4& Wei-Xing Zong1
Mounting evidence indicates that oncogenic Ras can modulate cell autonomous inflammatory
cytokine production, although the underlying mechanism remains unclear. Here we show that
squamous cell carcinoma antigens 1 and 2 (SCCA1/2), members of the Serpin family of
serine/cysteine protease inhibitors, are transcriptionally upregulated by oncogenic Ras via
MAPK and the ETS family transcription factor PEA3. Increased SCCA expression leads to
inhibition of protein turnover, unfolded protein response, activation of NF-kB and is essential
for Ras-mediated cytokine production and tumour growth. Analysis of human colorectal and
pancreatic tumour samples reveals a positive correlation between Ras mutation, enhanced
SCCA expression and IL-6 expression. These results indicate that SCCA is a Ras-responsive
factor that plays an important role in Ras-associated cytokine production and tumorigenesis.
DOI: 10.1038/ncomms4729
1Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA. 2Department of Pathology, Microbiology
and Immunology, Vanderbilt University, Nashville, Tennessee 37232, USA. 3Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, New York
11797, USA. 4Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224, USA. *These authors contributed equally to this work. Correspondence
and requests for materials should be addressed to W.-X.Z. (email: weixing.zong@stonybrook.edu).
NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729| www.nature.com/naturecommunications 1
&2014 Macmillan Publishers Limited. All rights reserved.
Constitutively active mutants of Ras are found in a large
number of human cancers1,2. Among the oncogenic
activities of mutant Ras, its ability to alter the tumour
microenvironment has been well appreciated with numerous
studies implicating Ras in various non-cell autonomous processes
including basement membrane degradation, immune cell
infiltration and angiogenesis3,4. This tumour microenvironment
altering function of Ras is at least partially caused by the ability of
Ras to induce the production and secretion of proinflammatory
and pro-tumorigenic cytokines. As cellular senescence has been
largely attributed to the cytokine secretory response termed
senescence-associated secretion phenotype (SASP)5,6, the
possibility remains that oncogene-induced secretory profile is a
senescence-independent process on Ras activation. Several
reports have implicated Ras in the ability to modulate the
tumour microenvironment at least due in part to Ras’ ability to
promote the production of cytokines and chemokines such as
Interleukin (IL)-6, IL-8, granulocyte–macrophage-colony-
stimulating factor (GM-CSF) in a cell autonomous
manner3,4,7,8. Therefore, it remains to be determined whether
premature senescence is a prerequisite of oncogene-induced
secretory phenotype, and how Ras mechanistically regulates the
expression of the proinflammatory cytokines.
Squamous cell carcinoma antigens (SCCAs) are members
of the serpin family of endogenous protease inhibitors.
Approximately 98 and 92% homologous at their nucleotide and
amino-acid levels, respectively, SCCA1 (SerpinB3) and SCCA2
(SerpinB4) are ‘suicide-substrate’ protease inhibitors with differ-
ing substrate specificities because of amino-acid differences
within their reactive site loop domain9. Upregulated in
numerous cancers (the cervical, lung, head and neck, liver and
breast)10–13, both SCCA1 and SCCA2 are thought to promote cell
survival through the inhibition of cell death14,15. Moreover, the
level of SCCA has been shown to predict pathological grade,
disease stage, recurrence and response to both radiotherapy and
chemotherapy16–18. Despite SCCA’s well-reported involvement in
cancer, how SCCA is transcriptionally upregulated during
transformation and contributes to tumour development remains
largely unknown. Here, we study the oncogenic regulation of
SCCA and uncover a novel proinflammatory role for SCCA
downstream of mutant Ras. We demonstrate that through MAPK
signalling and the ETS transcription factor PEA3, oncogenic Ras
upregulates the expression of SCCA1/2 (SerpinB3/B4) to promote
inflammatory cytokine production and xenograft tumour growth.
Moreover, SCCA upregulation is observed in human colorectal
and pancreatic cancer. Our findings suggest an important role of
serpins in Ras-driven tumorigenesis.
Results
Oncogenic Ras upregulates SCCA expression. Elevated
expression of SCCA has been found in numerous human
cancers. However, the underlying molecular mechanism of its
upregulation remains unclear. We began to study this by
expressing several oncoproteins (HRasV12, myristolated-Akt
(myr-Akt), and c-Myc) in IMR90 primary human lung fibro-
blasts. While myr-Akt and c-Myc failed to induce SCCA
expression, HRasV12 led to a marked increase in SCCA protein
levels (Fig. 1a). The SCCA antibody utilized was unable to
distinguish between the SCCA isoforms10, although quantitative
reverse-transcription PCR (qRT–PCR) analysis revealed an
increase in the transcript levels of both SCCA1 and SCCA2 in
response to RasV12 expression (Fig. 1b). This is not surprising
as the two SCCA isoforms are tandemly arranged on
human chromosome 18q and their promoters are highly
homologous19.
64 kDa Akt
Myc
Ras
SCCA
SCCA
Vector
HRas
Vector
HRas
Vector
KRas
Vector
KRas
Vector
KRas
β-Tubulin
β-Tubulin
Vector
myr-Akt
c-Myc
HRas
Vector
NRas
HRas
KRas
Ras
SCCA
β-Tubulin
Ras
64 kDa
22 kDa
22 kDa 22 kDa
IMR90 BJ HT-29 Caco-2 HeLa
ERK1/2
P-ERK1/2
:P-ERK/T-ERK
β-Tubulin
50 kDa
50 kDa
50 kDa
50 kDa
64 kDa
***
*
Vector
Ras
10,000
1,000
100
10
1
0.1
Relative expression
4-OHT: – ++
1.0
1.0 3.7 4.2 2.9
1.9 0.1
SCCA
ER:Ras
Day 12
Day 8
Day 0
:SCCA/tubulin
50 kDa
50 kDa 50 kDa
50 kDa
50 kDa 50 kDa
ER:Ras Day 0
ER:Ras+OHT Day 8
ER:Ras+OHT Day 12
ER:Ras-withdraw
***
1.2
1.0
0.8
Relative expression
0.6
0.4
0.2
0SCCA1 SCCA2
SCCA1 SCCA2
***
Figure 1 | Oncogenic Ras upregulates SCCA expression. (a) Indicated oncogenes were stably expressed in IMR90 cells. Whole-cell lysates were obtained
and analysed by western blot with indicated antibodies. (b) Total RNA was extracted from vector-control or RasV12-expressing IMR90 cells. SCCA1
and SCCA2 transcript levels were analysed via qRT–PCR and normalized to that in vector-control cells. Data shown are mean þs.e.m. of three independent
experiments performed in triplicate. (c,d) IMR90 cells expressing the ER:RasV12 fusion protein were treated with 4-OHT for 8 days, split and either cultured
in media containing 4-OHT or withdrew 4-OHT for additional 4 days. (c) Whole-cell lysates were analysed by western blot with indicated antibodies.
(d) Total RNA was extracted, and SCCA1 and SCCA2 transcript levels were analysed via qRT–PCR, and normalized to that of Day 12 ER:Ras with
4-OHT cells. Data shown are mean þs.e.m. of three independent experiments performed in triplicate. (e) Indicated oncogenic Ras proteins were
stably expressed in IMR90 cells. Whole-cell lysates were obtained and analysed by western blot with indicated antibodies. (f) HRasV12 was stably
expressed in IMR90 and BJ cell lines. KRasV12 was stably expressed in HT-29, Caco-2 and HeLa cells. Whole-cell lysates were obtained and analysed
by western blot with indicated antibodies. *Po0.05; ***Po0.001 by t-test.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729
2NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications
&2014 Macmillan Publishers Limited. All rights reserved.
To determine whether persistent Ras signalling is required for
SCCA expression, we used the oestrogen receptor (ER):RasV12
fusion protein that allows for RasV12 stabilization on the addition
of 4-hydroxytamoxifen (4-OHT) to the culture medium20. While
4-OHT induced the expression of Ras and SCCA (Fig. 1c),
removal of 4-OHT led to a drastic reduction in RasV12 protein
levels and diminished downstream signalling indicated by
decreased phospho-ERK, accompanied by a reduction of SCCA
at both protein and transcript levels (Fig. 1c,d), indicating that
sustained Ras signalling is required for maintaining SCCA
expression.
In addition to HRasV12, expression of KRasV12 or NRasQ61 also
resulted in elevated SCCA expression in IMR90 cells (Fig. 1e).
Similar to IMR90 cells, increased SCCA expression was observed
by expressing HRasV12 in the BJ primary human foreskin
fibroblast cells, and by expression of KRasV12 in a number of
cancer cell lines with wild-type Ras, including the colon cancer
cell lines HT-29 and Caco-2, and HeLa cells (Fig. 1f). Taken
together, these results indicate that upregulation of SCCA
expression is a general feature of oncogenic Ras proteins in
various human cell lines.
It is important to note that in IMR90 and BJ fibroblasts
expression of RasV12 elicits oncogene-induced senescence21. The
upregulation of SCCA in response to RasV12 in Caco-2, HT-29
and HeLa cells, which do not undergo senescence3,22, suggests
that upregulation of SCCA by RasV12 is independent of cellular
senescence. To further elucidate that the increased SCCA
expression is not a consequence of cellular senescence, we
induced premature senescence by treating IMR90 cells with
etoposide or H
2
O
2
and replicative senescence by continually
passaging the cells. Indeed, unlike RasV12, these senescence-
inducing conditions failed to upregulate SCCA expression
(Fig. 2). These results indicate that expression of oncogenic Ras
stimulates expression of SCCA in a manner that is independent of
oncogene-induced senescence.
Ras-induced SCCA is dependent on the MAPK/PEA3 pathway.
To elucidate the mechanism by which RasV12 stimulates SCCA
expression, we used pharmacological inhibitors to selectively
inhibit MAPK and Akt pathways that are well characterized to be
activated by Ras2,23. While inhibition of Akt signalling had
little to no effect on SCCA expression (Fig. 3a,b), the MEK
inhibitor U0126 caused a marked inhibition of both SCCA1 and
SCCA2 transcription in both IMR90 (Fig. 3c,d) and HeLa
cells (Supplementary Fig. 1a,b). Similarly, expression of the
dominant-negative ERK2 blocked Ras-induced SCCA expression
(Supplementary Fig. 1c,d). Along the Ras/Raf/MAPK/ERK
pathway, expression of oncogenic BRafV600E also induced
SCCA expression (Supplementary Fig. 1e). We next sought to
identify which transcription factor may be mediating Ras-induced
expression of SCCA. The ETS transcription factor family member
PEA3 has been shown to be modulated by MAPK via
sumolyation24 and can activate SCCA transcription25. Indeed,
silencing of PEA3 in RasV12-expressing cells (Fig. 3e) resulted in a
drastic decrease of both SCCA1 and SCCA2 at protein and
transcript levels (Fig. 3f,g), without affecting MAPK signalling
indicated by similar levels of phospho-ERK in RasV12-expressing
cells (Fig. 3f). This PEA3-dependent SCCA expression was also
observed in RasV12-expressing HeLa cells (Supplementary
Vehicle Etoposide
H2O2Ras
Early passage Late passage
Veh
Eto
H2O2
Ras
22 kDa
50 kDa
22 kDa
50 kDa β-Tubulin
p21
SCCA
Ras
EP
LP
Vector
Ras
22 kDa
50 kDa
22 kDa
50 kDa β-Tubulin
p21
SCCA
Ras
Figure 2 | DNA damage-induced and replicative senescence fails to upregulate SCCA. (a,b) IMR90 cells were treated with vehicle-control, etoposide
(10 mM) for 48 h, H
2
O
2
(10 mM) for 1 h, or stably transduced with HRasV12, then analysed 7 days post-treatment. (a) Cells were stained for b-Gal activity.
Representative images are shown. (b) Whole-cell lysates were analysed by western blot with indicated antibodies. (c,d) IMR90 cells were continuously
passaged and harvested at passage 15 as early passage (EP) or at passage 30 as late passage. (c) Cells were stained for b-galactosidase activity.
Representative images are shown. (d) Whole-cell lysates were analysed by western blot with indicated antibodies. Note that while all the conditions induce
cellular senescence, only RasV12 led to SCCA expression. Scale bar ¼100 mm.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729 ARTICLE
NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications 3
&2014 Macmillan Publishers Limited. All rights reserved.
Fig. 2a–c) and was rescued by expression of wild-type murine
PEA3, but not by a PEA3 sumolyation-defective mutant24
(Supplementary Fig. 2d–f). These results indicate that mutant
Ras leads to SCCA upregulation via the MAPK/PEA3 pathway.
SCCA is essential for Ras-induced cytokine production. Next
we examined the biological role of elevated SCCA expression. We
chose to focus on using the IMR90 cell line that is widely used for
studying the effect of Ras activation during the early phase of
tumorigenesis. We began by examining the downstream signal-
ling pathways activated by oncogenic Ras (MAPK, Akt, NF-kB).
To this end, SCCA expression was silenced in RasV12-expressing
cells using two independent shRNA constructs that effectively
silenced both SCCA1 and SCCA2 (Fig. 4a), and the signalling
pathways examined by respective phosphorylation antibodies.
While loss of SCCA had little to no effect on the ability of mutant
Ras to activate Akt and MAPK phosphorylation (Fig. 4b), loss of
SCCA significantly diminished RasV12-induced RelA/p65 phos-
phorylation that is indicative of NF-kB activation (Fig. 4b). This
effect was further confirmed by utilizing a NF-kB luciferase
reporter construct (Fig. 4c).
Activation of NF-kB signalling has been attributed to cytokine
production downstream of Ras activation26. Indeed, inhibition
of NF-kB signalling using the pharmacological inhibitor BAY11-
7082 abrogated Ras-induced cytokine production in IMR90 and
HeLa cells (Supplementary Fig. 3). Therefore, we examined
whether SCCA plays a role in Ras-induced cytokine production
and performed a quantitative cytokine array analysis using cell
culture media. In agreement with literature5,6, RasV12-expressing
cells displayed a marked increase in a spectrum of cytokines
including IL-6, IL-8, CXCL1, G-CSF and GM-CSF (Fig. 4d).
Vector Ras
1.6
1.4
1.2
1.0
0.2
0.4
0.6
0.8
0
SCCA1 SCCA2
Vector Ras
Ras
SCCA
++
AKTi:
22 kDa
50 kDa
1.0 0.9
Ras
SCCA
:SCCA/tubulin
P-Akt (Ser473)
:P-Akt/T-Akt
β-Tubulin
0.72.10.3
50 kDa
50 kDa
22 kDa
MEKi:
64 kDa
64 kDa
1.0
Akt
++
P-ERK1/2
:SCCA/tubulin
ERK1/2
β-Tubulin
shNTC
shPEA3 ***
3.0
2.5
2.0
1.5
1.0
0.5
0Vector Ras
Relative expression
:P-ERK/T-ERK
1.4 0.60.3
0.3
1.0
1.0
50 kDa
50 kDa
50 kDa
Vector Ras
Ras
SCCA
50 kDa
22 kDa
shPEA3
shNTC
shREA3
shNTC
P-ERK1/2
:SCCA/tubulin
ERK1/2
β-Tubulin
:P-ERK/T-ERK1.9 2.11.0
0.5
1.0
1.0
50 kDa
50 kDa
50 kDa
NS
NS
Vector + vehicle
Vector + AKTi
Ras + vehicle
Ras + AKTi
Relative expression
1.2
1.0
0.2
0.4
0.6
0.8
0
SCCA1 SCCA2
**
**
Vector + vehicle
Vector + MEKi
Ras + vehicle
Ras + MEKi
Relative expression
1.2
1.0
0.2
0.4
0.6
0.8
0
SCCA1 SCCA2
***
***
Vector-shNTC
Vector-shPEA3
Ras-shNTC
Ras-shPEA3
Relative expression
Figure 3 | SCCA upregulation is mediated by MAPK/PEA3. (a,b) Vector-control or HRasV12-expressing IMR90 cells were treated with either
vehicle-control or AKTi (10 mM) for 24 h. (a) Whole-cell lysates were analysed by western blot with indicated antibodies. (b) Total RNA was extracted
and SCCA1 and SCCA2 transcript levels were analysed via qRT–PCR, and normalized to RasV12 cells treated with vehicle-control. Data shown are
mean þs.e.m. of two independent experiments performed in triplicate. (c,d) Vector-control or HRasV12-expressing IMR90 cells were treated with
either vehicle-control (DMSO) or U1026 (MEKi, 10mM) for 24 h. (c) Whole-cell lysates were analysed by western blot with indicated antibodies.
(d) Total RNA was extracted, and SCCA1 and SCCA2 transcript levels were analysed via qRT–PCR and normalized to that of Ras-expressing cells
treated with vehicle. Data shown are mean þs.e.m. of three independent experiments performed in triplicate. (eg) Vector-control or HRasV12-expressing
IMR90 cells were stably infected with lentiviral shNTC (non-target control) or shPEA3. (e) Successful silencing of PEA3 was confirmed via qRT–PCR
and normalized to that in vector-control cells with shNTC. Data shown are mean þs.e.m. of three independent experiments performed in triplicate.
(f) Whole-cell lysates were analysed through western blot with indicated antibodies. (g) Total RNA was analysed for the transcript levels of SCCA1
and SCCA2 via qRT–PCR and normalized to that in Ras-expressing cells with shNTC. Data shown are meanþs.e.m. of three independent experiments
performed in triplicate. **Po0.01; ***Po0.001; NS, non-significant by t-test.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729
4NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications
&2014 Macmillan Publishers Limited. All rights reserved.
Consistent with the observed decrease in NF-kB signalling
(Fig. 4b,c), this increased cytokine expression was significantly
abrogated on SCCA silencing (Fig. 4d,e). Quantitative RT-PCR
analysis revealed that the suppression of cytokine production was
due to transcriptional downregulation (Fig. 4f) and was rescued
by ectopically activating NF-kB signalling with TNFatreatment
(Fig. 4g). The SCCA-dependent NF-kB activation and cytokine
production downstream of oncogenic Ras were also observed in
HeLa and Caco-2 cells (Supplementary Fig. 4). The introduction
of oncogenic Ras in IMR90 elicits a DNA damage response that
has been reported to mediate cytokine production6,27. However,
silencing of SCCA had virtually no effect on Ras-induced DNA
damage indicated by the phosphorylation of H2A.X (Fig. 5a,b)
or the senescence-associated cell cycle arrest (Fig. 5c).
Taken together, these results indicate that SCCA plays an
essential role in Ras-mediated NF-kB activation and
inflammatory cytokine production that is independent of the
DNA-damage response.
GM-CSF
G-CSF
CXCL1
IL-8
IL-6
shSCCA#2
shSCCA#1
shNTC
Vector
***
***
1.2
1.0
0.8
Relative expression
0.6
0.4
0.2
0
IL-6 IL-8 CXCL1 G-CSF GM-CSF
1.2
1.0
0.8
Relative amount of
secreted protein
0.6
0.4
0.2
0IL-6 IL-8 CXCL1 G-CSF GM-CSF
***
*** ***
*** ***
*** ***
***
Vector
***
***
25
20
15
10
5
0
Relative luminescence
(NF-kB activity)
Ras-shNTC
Ras-shSCCA#1
Ras-shSCCA#2
***
***
***
**
*
4.0
3.0
2.0
Relative expression
1.0
0
IL-6 IL-8 CXCL1 G-CSF GM-CSF
Ras
Ras
64 kDa
64 kDa
50 kDa
50 kDa
50 kDa
64 kDa
64 kDa
1.0 3.7 3.0 4.2
P-Akt (Ser473)
Akt
:P-Akt/T-Akt
:P-ERK/T-ERK
P-RelA/p65
RelA/p65
:P-RelA/T-RelA2.42.13.81.0
1.81.92.01.0
β-Tubulin
P-ERK1/2
ERK1/2
22 kDa
50 kDa
50 kDa
Ras
Vector
shNTC
shSCCA#1
shSCCA#2
Vector
shNTC
shSCCA#1
shSCCA#2
SCCA
β-Tubulin
Ras
ab
d
fg
e
c
Vector
Ras-shNTC
Ras-shSCCA
Ras-shSCCA+TNFa
Vector
Ras-shNTC
Ras-shSCCA#1
Ras-shSCCA#2
Vector
Ras-shNTC
Ras-shSCCA#1
Ras-shSCCA#2
Figure 4 | SCCA modulates Ras-induced cytokine production. (af) Vector-control or HRasV12-expressing IMR90 cells were stably infected with lentiviral
shRNA control (shNTC) or two independent hairpins targeting SCCA. (a,b) Whole-cell lysates were analysed by western blot with indicated antibodies.
(c) Cells were transfected with an NF-kB luciferase reporter and a renilla luciferase construct. Twenty-four hours post transfection, cells were lysed
and luminescence was quantified. NF-kB luciferase activity was standardized based on renilla luciferase activity and normalized to that of vector-control
cells. Data shown are mean þs.e.m. of three independent experiments performed in triplicate. (d) Culture media were collected and subjected to a cytokine
antibody array. The representative of two independent blots of indicated cytokines are shown. (e) The relative amount of cytokines was quantified and
normalized to that of RasV12-shNTC cells. (f) Total RNA was extracted and cytokine transcript levels were analysed via qRT–PCR, and normalized to
that of RasV12-shNTC cells. Data shown are mean þs.e.m. of three independent experiments performed in triplicate. (g) Ras-shSCCA cells were
treated with TNFa(50 ng ml1) for 16 h. Total RNA was extracted and cytokine transcript levels were analysed via qRT–PCR and normalized to that of
RasV12-shNTC cells. Data shown are mean þs.e.m. of two independent experiments performed in triplicate. *Po0.05; **Po0.01; ***Po0.001 by t-test.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729 ARTICLE
NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications 5
&2014 Macmillan Publishers Limited. All rights reserved.
SCCA promotes cytokine production by inducing the unfolded
protein response. We next sought to understand how SCCA
promotes Ras-mediated NF-kB activation and cytokine produc-
tion. A possible mechanism is through upregulation of the
unfolded protein response (UPR), which has been implicated in
mediating NF-kB activation and inflammatory transcriptional
programmes28. Our previous work showed that SCCA leads to
decreased lysosomal and proteasomal protein turnover, two
major protein degradation pathways, and hence increases the
steady-state level of ER stress29. Thus, we speculated that mutant
Ras induces ER stress through upregulation of SCCA. Indeed,
the ability of RasV12 to promote an ER stress response has
been reported30. Silencing of SCCA relieved the Ras-induced
inhibition of protein turnover as measured through total
ubiquitinated proteins (Fig. 6a). Furthermore, while RasV12 cells
showed a robust increase in UPR indicated by the appearance of
proteolytically cleaved ATF6 as well as both unspliced and spliced
XBP1, this response was significantly reduced in SCCA-silenced
cells (Fig. 6b), indicating that SCCA mediates the UPR induced
by RasV12. Silencing of ATF6 or XBP1 in RasV12-expressing
cells (Fig. 6c) drastically diminished RasV12-induced cytokine
production (Fig. 6d–f; Supplementary Fig. 5) that was
accompanied by decreased RelA/p65 phosphorylation (Fig. 6g;
Supplementary Fig. 5). Conversely, ectopic expression of the
transcriptionally active spliced variant of XBP1 (XBP1s) restored
cytokine production in RasV12 cells when SCCA was silenced
(Fig. 6h,i). These results indicate that SCCA modulates cytokine
production downstream of mutant Ras by stimulating UPR.
SCCA is upregulated in colorectal and pancreatic cancers. Our
results thus far show that SCCA is a Ras-responsive gene that
plays an essential role in Ras-induced cytokine production. We
next sought to identify an in vivo connection between Ras
and SCCA expression. Using TCGA (The Cancer Genome Atlas)
data of human colorectal cancer, which were available with
sizeable samples with the KRas mutation, we identified a positive
correlation between the presence of mutant Ras and SCCA
upregulation (Fig. 7a). We also chose to examine pancreatic
ductal adenocarcinoma (PDAC), where the mutant KRas func-
tions as the primary driver31. It also offers a unique model where
the precursor lesions to PDAC have a defined pathological
progression31. To this end, we analysed the pancreatic cancer
gene expression data sets for SCCA expression through
Oncomine. In the seven available patient data sets, SCCA1
mRNA was upregulated in six and SCCA2 mRNA in three
independent data sets32–37 (Fig. 7b–d). Of note, the Logsdon data
set37 identified both SCCA1 and SCCA2 to be upregulated in
pancreatic cancer when compared with chronic pancreatitis
(Fig. 7d). To further examine the association of SCCA expression
in human pancreatic cancer, we performed immunohisto-
chemistry (IHC) for SCCA using a pancreatic cancer tissue
microarray. While all non-neoplastic/normal pancreatic samples
were negative for SCCA expression, positive staining for SCCA
was seen throughout all stages of pancreatic cancer progression.
The incidence of SCCA positivity increased in relation to the
progression of pancreatic cancer: 2 out of 17 (11.8%) PanIN-1
lesions, 5 out of 19 (26.3%) PanIN-2 lesions, 8 out of 15 (53.3%)
PanIN-3 lesions and 20 out of 30 (66.7%) of PDAC (Fig. 7e;
Po0.001). Moreover, consistent with the result that SCCA
promotes inflammatory cytokine production in cultured cells,
IHC of IL-6 on the same panel of pancreatic tissues showed a
positive correlation between IL-6 and SCCA in both PDAC
(Fig. 7f,g) and PanIN (Supplementary Fig. 6) samples.
Endogenous SCCA promotes IL-6 production and tumor-
igenesis. To further explore the physiological relevance of SCCA,
we tested the expression and function of SCCA in a setting when
mutant Ras is expressed at endogenous levels. RasV12 failed to
induce the expression of the SCCA paralogues Serpinb3a or Ser-
pinb3b in immortalized MEF NIH3T3 (Supplementary Fig. 7a) or
in primary MEFs (Supplementary Fig. 7b). The failure to detect
upregulation of SCCA paralogues in the murine system prevented
us from using the well-established LSL-KRas mouse model38.
Hence, we instead examined a panel of human pancreatic cancer
cell lines that harbour the mutant KRas. We found five out of nine
cell lines to be positive for SCCA expression (Fig. 8a). In the cell
lines that displayed detectable levels of SCCA and can tolerate loss
of KRas (CFPAC-1, L3.6 and Capan-1), silencing of mutant KRas
significantly reduced SCCA expression (Fig. 8b). Consistent with
the essential role of SCCA in Ras-mediated cytokine production,
silencing of SCCA in three SCCA-positive cell lines resulted in
decreased expression of IL-6, whereas the SCCA shRNAs did not
affect IL-6 expression in the SCCA-low PANC-1 cells (Fig. 8c).
Moreover, while silencing of SCCA in CFPAC-1, L3.6 and HPAF-II
cells did not compromise cell proliferation in cell culture
(Supplementary Fig. 8), it led to impaired xenograft tumour
growth in all three cell lines (Fig. 8d). These results, taken together
with the above finding that SCCA positivity correlates with
progression of pancreatic cancer development (Fig. 7e), indicate
that SCCA expression is under the control of mutant Ras expressed
at endogenous level, and that the SCCA-mediated inflammatory
response plays a pro-tumorigenic role in Ras-driven cancer.
DAPI
Vector
Ras-shNTC
Ras-shSCCA#1
Ras-shSCCA#2
γH2AX Merge Vector
Ras-shNTC
Ras-shSCCA#1
NS
35
30
25
20
15
% γH2AX-positive cells
10
5
0
35
45
55
BrdU
SAHF
NS
Vector
Ras
shNTC
Ras
shSCCA#1
Ras
shSCCA#2
25
15
% Positive cells
5
–5
Ras-shSCCA#2
Figure 5 | Silencing of SCCA does not interfere with the DNA-damage response. (ac) Vector-control or HRasV12-expressing IMR90 cells were stably
infected with lentiviral shRNA control (shNTC) or two independent hairpins targeting SCCA (same cells as shown in Fig. 4). (a) Immunofluorescence
against gH2A.X was performed. Representative images are shown; scale bar, 20 mm. (b) Quantification of percent gH2A.X-positive cells is shown. Note that
silencing of SCCA does not compromise Ras-induced DNA damage. Data shown are mean þs.e.m. of three independent experiments. (c) Cells were
cultured with BrdU (10 mM) for 6 h and immunofluorescence against BrdU was performed. Quantification of BrdU-positive and senescence-associated
heterochromatic foci (SAHF)-positive cells are shown. Data shown are mean þs.e.m. of three independent experiments. NS, non-significant by t-test.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729
6NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications
&2014 Macmillan Publishers Limited. All rights reserved.
Discussion
In this study, we show that the endogenous serine/cysteine
proteinase inhibitor SCCA is upregulated on activation of
oncogenic Ras, in a manner that is dependent on hyper-active
MAPK signalling and the ETS transcription factor PEA3.
Downstream of oncogenic Ras, SCCA promotes activation of
NF-kB signalling and cytokine production by blocking protein
turnover and inducing the UPR.
Our study implicates SCCA as a direct link between oncogenic
Ras mutation, NF-kB activation and cytokine production.
Vector
shNTC
shSCCA#1
shSCCA#2
Ras
Vector
shNTC
shSCCA#1
shSCCA#2
Ras
Vector
shNTC
shATF6
shXBP1
Ras
Ras
β-Tubulin
RT-PCR
GAPDH
ATF6
XBP1
Ras
22 kDa
22 kDa
22 kDa
50 kDa
96 kDa
36 kDa
50 kDa
50 kDa
50 kDa
Ras
Vector
shNTC
shATF6
shXBP1
β-Tubulin
:XBP1u/tubulin
:XBP1s/tubulin
:ATF6/tubulin1.0
1.0 2.2 1.2
5.4 3.6 4.0
1.0
1.0 2.6 1.0 0.3
ATF6 p50
ATF6 p90
Ras
XBP1u
XBP1s
96 kDa
64 kDa
50 kDa
50 kDa
1.0 4.0 1.6 1.7
Ubiquitin
:Ubiquitin/tubulin
β-Tubulin
IL-6
IL-8
CXCL1
G-CSF
GM-CSF 0
IL-6 IL-8 CXCL1 G-CSF GM-CSF
0.2
0.4
0.6
Relative amount of
secreted protein
Relative expression
0.8
1.0
0
Ras – +
++
++
++
+
+
Ras
SCCA
1.0
1.0 8.7
53 2.0 6.2
4.3 10
:SCCA/tubulin
XBP1s
:XBP1s/tubulin
β-Tubulin
++
–––
shNTC
shSCCA
Vector
XBP1s
22 kDa
50 kDa
50 kDa
50 kDa
0.2
0.4
0.6
0.8
1.0
1.2
Vector
Ras-shNTC
Ras-shATF6
Ras-shXBP1
0IL-6 IL-8 CXCL1 G-CSF GM-CSF
0.2
0.4
0.6
Relative expression
0.8
1.0
1.2
Vector
Ras-shNTC
Ras-shSCCA
Ras-shSCCA-XBP1s
***
***
***
NS
**
Vector
Ras-shNTC
Ras-shATF6
Ras-shXBP1
***
*** ***
*** ***
*** ***
*** ***
64 kDa
64 kDa
50 kDa
1.0 2.0 1.0 1.4
Ras
Vector
shNTC
shATF6
shXBP1
β-Tubulin
P-RelA/p65
:P-RelA/T-RelA
RelA/p65
***
1.2
IL-6 IL-8 CXCL1 G-CSF GM-CSF
Figure 6 | SCCA promotes cytokine production by inducing UPR. (a,b) Vector-control or HRasV12-expressing IMR90 cells were stably infected with
lentiviral shRNA control (shNTC) or two independent hairpins targeting SCCA. Whole-cell lysates were analysed by western blot with indicated antibodies.
Quantification of the ubiquitin blot in awas performed by Image J using the full-length of each lane. (cg) Vector-control or HRasV12 IMR90 cells
were stably infected with lentiviral shRNA control (shNTC) or shRNA hairpins targeting ATF6 or XBP1. (c,g) Whole-cell lysates were analysed by western
blot with indicated antibodies. (d) Culture media were collected and subjected to a cytokine antibody array. The blots of indicated cytokines are shown.
(e) The relative amount of cytokines was quantified and normalized to that of RasV12-shNTC cells. (f) Total RNA was extracted and cytokine transcript
levels were analysed via qRT–PCR and normalized against that in RasV12-shNTC cells. Data shown are mean þs.e.m. of three independent experiments
performed in triplicate. (h,i) Vector-control or HRasV12 IMR90 cells were stably infected with lentiviral shRNA control (shNTC) or shSCCA, together
with vector-control or XBP1s-expressing construct. (h) Whole-cell lysates were analysed by western blot with indicated antibodies. (i) Total RNA was
extracted and cytokine transcript levels were analysed via qRT–PCR and normalized against that in RasV12-shNTC cells. Data shown are mean þs.e.m.
of three independent experiments performed in triplicate. **Po0.01; ***Po0.001; NS, non-significant by t-test.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729 ARTICLE
NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications 7
&2014 Macmillan Publishers Limited. All rights reserved.
While it has been clearly demonstrated that the mutant Ras can
activate NF-kB and induce inflammatory cytokine production in
various settings, the underlying molecular mechanism has
remained elusive. In numerous settings, inflammatory cytokine
production has been attributed to oncogene-induced senescence
that is associated with excess production of reactive oxygen
species and subsequent DNA damage or loss of p53 (refs 6,27).
We found that oncogenic Ras upregulates SCCA to promote
TCGA colorectal
Wild-type
KRas
Mutant
KRas
P= 0.012
Pei pancreas
4
2
0
–2
–1.5
–3.0
–1.5
1.5
3.0
0
1.5
3.0
0
4
6
2
0
–2
P= 3.95E-7
P= 8.85E-6 P= 7.51E-4 P= 0.002
–1.5
1.5
3.0
0
P= 0.050
–4
–2
0
2
4
Pei pancreas Badea pancreas SCCA1
Logsdon pancreas
SCCA2
Logsdon pancreas
Normal
Cancer
Normal
SCCA
SCCA
SCCA negative SCCA positive
100
80
60
40
% Cases
20
0
SCCA
negative
SCCA
positive
P= 0.0385
High
Moderate
Low
Negative
IL-6
PanIN-1 PanIN-2 PanIN-3 PDAC
P= 1.03E-5
1.5
3.0
0
–1.5
–3
–2
–1
0
–6
–2
–4
0
2
P= 7.37E-4 P= 0.006 P=0.043
Normal
Cancer
Cancer
Pancreatitis
Badea pancreas Iacoduzio-Donahue
pancreas
Segara
pancreas
Grutzmann
pancreas
mRNA expression
(log2 intensity)
mRNA expression
(log2 intensity)
mRNA expression
(log2 intensity)
mRNA expression
(log2 intensity)
–6
–4
–2
0
Figure 7 | SCCA is upregulated in human colorectal and pancreatic cancer. (a) TCGA human colorectal cancer data of somatic mutation and RNA
expression from Broad Institute’s Genome Data Analysis Center were analysed. There were 207 human colorectal tumours that have both somatic
mutation and mRNA expression data available. KRas was mutated in 87 out of the 207 samples. By comparing SCCA mRNA expression level of the groups
with wild-type and mutated KRas, SCCA expression was found to be significantly higher in the group with KRas mutation. Boxplots with whisker from 10 to
90 percentile are shown. SCCA expression log2 intensity values for wild-type (n¼120) and mutant (n¼87) KRas samples are shown. P¼0.012 by
Wilcoxon Rank Sum test. (bd) Oncomine (www.oncomine.org) data sets were analysed for SCCA1 (b) or SCCA2 (c) mRNA expression levels in normal
pancreatic tissue and pancreatic cancer, or for SCCA1 and SCCA2 mRNA expression levels in chronic pancreatitis and pancreatic cancer (d). The boxes
represent the interquartile range. Whiskers represent the 10th–90th percentile range. Bars represent the median. P-values were calculated by two-sample
t-test. (eg) IHC against SCCA and IL-6 was performed on the corresponding serial pancreatic tissue microarrays. (e) Representative images of
normal pancreatic tissue and SCCA-positive PanIN 1, PanIN-2, PanIN-3 and PDAC samples are shown; scale bar, 100 mm. (f) Representative images of
SCCA/IL-6-negative and SCCA/IL-6-positive grade III PDAC samples; scale bar, 50 mm. (g) Quantification of IL-6 staining in SCCA-negative and
SCCA-positive samples. w2-test for trend was used to determine significance, P¼0.0385.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729
8NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications
&2014 Macmillan Publishers Limited. All rights reserved.
cytokine production, whereas SCCA is not upregulated in the
setting of drug-induced and replicative senescence. These results
suggest that the secretory response seen in cells with oncogenic
Ras mutations may be distinct from SASP induced by DNA
damage or loss of p53. Although the physiological significance of
Ras-induced cellular senescence remains an open topic, our
results show that Ras can induce SCCA expression in cells where
Ras does not induce senescence (Fig. 1f). Furthermore, we found
that SCCA-dependent cytokine production occurs in human cells
expressing endogenous level of mutant Ras (Fig. 8) and this
provides a mechanism that uncouples cellular senescence
from inflammatory cytokine production on Ras activation.
Importantly, our study shows that the incidence of SCCA
positivity within PanIN lesions and PDAC increases with disease
progression (Fig. 7e). Therefore, monitoring SCCA levels may
become an attractive diagnostic tool in pancreatic cancer patients,
as the levels of SCCA have been used in numerous studies to
successfully predict disease stage and response to therapy16–18.
Our work uncovers SCCA as a critical mediator of Ras-induced
UPR. Until now, SCCA’s involvement in cancer was largely
attributed to its protective role against lysosomal permeability
that leads to unscheduled activation of lysosomal proteases29,39.
We have previously reported that, through its protease inhibitory
activity, SCCA leads to a decrease in both lysosomal and
proteasomal protein turnover and elicits a non-lethal level of ER
stress response29. It remains to be determined how SCCA
mechanistically suppresses protein turnover, and whether other
functions of SCCA such as its possible role in the nucleus
contribute to increased UPR. Nevertheless, here we show that
SCCA-mediated UPR plays a critical role in the context of
oncogenic Ras mutation. Enhanced UPR has been well implicated
in cancer, largely owing to the highly proliferative status of cancer
50 kDa
50 kDa
AsPC-1
Capan-1
Capan-2
CFPAC-1
HPAF-II
L3.6
PANC-1
PL45
S2-013
SCCA
22 kDa
shNTC
shKRas
shNTC
shKRas
shNTC
shKRas
50 kDa
50 kDa
1.2
1.0
0.8
Relative expression
0.6
0.4
0.2
0
CFPAC-1
50 kDa
50 kDa
***
*** *
*
IL-6
***
***
IL-6
NS
1.5
1.0
0.5
0IL-6IL-6
1.2
1.0
0.8
0.6
0.4
Relative
expression
0.2
0
L3.6
shNTC
shSCCA#1
shSCCA#2
shNTC
shSCCA#1
shSCCA#2
shNTC
shSCCA#1
shSCCA#2
shNTC
shNTC
shNTC
CFPAC-1
*
*
*
60 5 10 15 2050403020
Time post injection (days) Time post injection (days)
10 20 30
Time post injection (days)
10
0
50
100
150
200
250
300
350
200
200
300
400
100
00
400
600
800
1,000
1,200
Tumour volume (mm3)
shSCCA#1
shSCCA#2
shSCCA#2 shNTC
L3.6 HPAF-II **
*
*
shSCCA#1 *
*
shNTC
shSCCA#1
shSCCA#1
shSCCA#2
HPAF-II PANC-1
1.0 0.6
1.0 0.3
SCCA
KRas
CFPAC-1 L3.6 Capan-1
β-Tubulin
SCCA2SCCA1 SCCA2SCCA1 SCCA2SCCA1
1.0 0.3
1.0 0.3
1.0 0.5
1.0 0.5
SCCA
KRas
β-Tubulin
SCCA
:SCCA/tubulin
shNTC
shKRas
**
*********
*** *
:KRas/tubulin
KRas
β-Tubulin
β-Tubulin
SCCA
β-Tubulin
Figure 8 | Ras-dependent SCCA expression promotes IL-6 production and tumorigenesis in human pancreatic cancer cell lines. (a) Whole-cell lysates
from a panel of pancreatic cancer cells were obtained and analysed through western blot with indicated antibodies. (b) Indicated cell lines were stably
infected with lentiviral shNTC or shKRas. Whole-cell lysates were analysed through western blot with indicated antibodies. Note that the Ras antibody
utilized is a pan-Ras antibody and KRas is indicated by arrowhead. Total RNA was extracted, and SCCA1 and SCCA2 transcript levels were analysed via
qRT–PCR and normalized to that of shNTC cells. Data shown are mean þs.e.m. of three independent experiments performed in triplicate. (c) Indicated cell
lines were stably infected with lentiviral shNTC or shSCCA. Whole-cells lysates were analysed through western blot with indicated antibodies. Total RNA
was extracted and IL-6 transcript levels were analysed via qRT–PCR. Data shown are meanþs.e.m. of three independent experiments performed in
triplicate. Relative level of transcript was normalized to that of shNTC cells. Note that silencing of SCCA in PANC-1 cells, which have undetectable SCCA
expression, had virtually no effect on IL-6 production. (d) CFPAC-1, L3.6 and HPAF-II cells were injected into the flanks of athymic nude mice and monitored
for tumour growth; n¼5. Representative images of tumour and the tumour growth curve±s.e.m. are shown; scale bar, 1 cm. *Po0.05, **Po0.01,
***Po0.001 by t-test.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729 ARTICLE
NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications 9
&2014 Macmillan Publishers Limited. All rights reserved.
cells and their encounter of growth limiting conditions such as
nutrient deprivation and hypoxia40,41. Targeting the enhanced
UPR of cancer cells remains an attractive therapeutic option,
although the precise prediction of sensitivity has been difficult.
Consistent with this notion, tumours driven by the mutant Ras
have been shown to be susceptible to proteotoxic therapies42. Our
current work showing that SCCA expression is stimulated by
oncogenic Ras and plays an essential role in Ras-induced ER
stress response, together with our previous report demonstrating
SCCA sensitizes cells to proteotoxic stress29, suggest that SCCA
may be a valuable predictor of response to proteotoxic agents in
tumours harbouring oncogenic Ras.
The contribution of the tumour stroma and inflammation to
tumour development and progression, especially in pancreatic
and colorectal cancer, is well accepted43,44. While NF-kB
signalling has been shown to mediate the SASP response26,
it is critical for the development of Ras-driven lung
adenocarcinoma45. The pro-tumorigenic role of SCCA
mediating NF-kB activation and inflammatory cytokine
production is consistent with these reports. This adds a new
pro-tumour mechanism of SCCA in addition to its well-
documented anti-cell death function15,46–49. In a parallel study,
we find that ectopic expression of SCCA can independently
promote the transformation of the non-tumorigenic mammary
epithelial cell line MCF10A (Sheshadri et al., in preparation).
As numerous studies utilizing anti-inflammatories to target the
tumour microenvironment have shown great promise in the
treatment of malignancies50,51, the ability of SCCA to promote
cytokine production and tumorigenesis suggests a therapeutic
value of targeting its protease inhibitory activity.
Methods
Cell lines and culture.293T, IMR90, BJ, HT-29, Caco-2, HeLa, AsPC-1, Capan-1,
Capan-2, CFPAC-1, HPAF-II, L3.6, PANC-1, PL45 and S2-013 cells were cultured
according to ATCC recommendations. 293T, HeLa, PANC-1, PL45 and S2-013
cells were cultured in DMEM supplemented with 10% FBS (HyClone). IMR90,
BJ and HPAF-II cells were cultured in MEM supplemented with 10% FBS. HT-29
and Capan-2 cells were cultured in McCoy’s 5a supplemented with 10% FBS.
Caco-2 cells were cultured in MEM supplemented with 20% FBS. AsPC-1 and L3.6
cells were cultured in RPMI supplemented with 10% FBS. Capan-1 cells were
cultured in Iscove’s supplemented with 20% FBS. CFPAC-1 cells were cultured in
Iscove’s supplemented with 10% FBS. All media were supplemented with 100 units
per ml penicillin and 100 mgml1streptomycin (Invitrogen).
Plasmids and reagents.Retroviral expression vectors for WZL-hygro and
WZL-HRasV12 were a kind gift of Dr Alea Mills at Cold Spring Harbor
Laboratory21. pBABE-puro and pBABE-KRasV12 were a kind gift of Dr Scott Lowe
at Memorial Sloan-Kettering Cancer Institute. pBABE-NRasQ61 was purchased
from Addgene (Dr Channing Der; Plasmid 12543)52. pLNCX-ER:HRasV12 was a
kind gift of Dr Masashi Narita at University of Cambridge20. pcDNA3-FLAG-
ERK2-WT and pcDNA3-FLAG-ERK2-T138A (DN) were a kind gift of Dr Scott
Eblen at the Medical University of South Carolina53. pcDNA3-PEA3-WT and
pcDNA3-PEA3-K123R (MUT) were a kind gift of Dr Andrew Sharrocks at the
University of Manchester24. Human XBP1s was cloned by RT-PCR from total
RNA from IMR90 cells. Primers used were as follows, forward with HindIII
restriction and FLAG tag: 50-AAGCTTATGGATTACAAGGATGACGATGAC
AAGTGGTGGTGGCAGCCGCGCCGAACCC-30and reverse primer with HindIII
restriction site: 50-AAGCTTTTAGACACTAATCAGCTGGGGAAAG-30. The RT-
PCR product was digested with HindIII and ligated into the pLPC retroviral
expression vector. All shRNA lentiviral constructs were in the pLKO (Sigma)
backbone. shRNA targeting sequences used are shGFP: 50-TACAACAG
CCACAACGTCTAT-30; shScramble: 50-CAACAAGATGAAGAGCACCAA-30;
shSCCA#1: 50-GCACAACAGATTAAGAAGGTT-30; shSCCA#2: 50-CCGCTGTA
GTAGGGATTCGGAT-30; shPEA3: 50-GCTCCGATACTATTATGAGAA-30;
shATF6: 50-GCAGCAACCAATTATCAGTTT-30; shXBP1: 50-GCCTGTCTGTA
CTTCCATTCAA-30; shKRas: 50-CAGTTGAGACCTTCTAATTGG-30. Bay-11-
7082 (B5556), 4-hydroxytamoxifen (H7904) and crystal violet (C0775) were
purchased from Sigma. U0126 (V1121) was purchased from Promega. AKT
inhibitor VIII (124018) was purchased from Calbiochem. TNFa(210-TA) was
purchased from R&D Systems. X-gal (15520-018) was purchased from Invitrogen.
Etoposide (8154-1) was purchased from Clontech.
DNA transfection and viral infection.Both retrovirus and shRNA lentivirus were
generated in 293T cells. In brief, 293T were transfected by Lipofectamine 2000
(Invitrogen) with the plasmid of interest, packaging plasmid, and a plasmid
encoding for the VSV-G envelope protein. Forty-eight and seventy-two hours after
initial transfection, viral supernatant was collected, filtered, supplemented with
polybrene (10 mgml1) and used to infect target cells. Forty-eight hours after last
infection, cells were selected with appropriate antibiotics. IMR90 cells were selected
for 2 days with 100 mgml1of hygromycin, 2 mgml1of puromycin or
1.75 mg ml 1of G418. HeLa and L3.6 cells were selected with 0.5 mgml1of
puromycin for 2 days. Caco-2, Capan-1, CFPAC-1 and PANC-1 cells were selected
with 1.0 mgml1of puromycin for 3 days.
Immunoblot analysis.Cell lysates were prepared in RIPA buffer (1% sodium
deoxycholate, 0.1% SDS, 1% Triton X-100, 0.01 M Tris pH 8.0, 0.14 M NaCl).
Protein expression was examined by western blotting using antibodies against
SCCA1/2 (FL-390; Santa Cruz; 1:1,000), Ras (Clone Ras10; Millipore; 1:10,000),
p21 (C-19; Santa Cruz; 1:500), ERK1/2 (4,695; Cell Signaling; 1:10,000), P-ERK1/2
(4,370; Cell Signaling; 1:2;000), Akt (9,272; Cell Signaling; 1;2,000), P-Akt (4,058;
Cell Signaling; 1:2,000), P-RelA/p65 (3,031; Cell Signaling; 1;1,000), RelA/p65 (F-6;
Santa Cruz; 1:2,000), ATF6 (F-7; Santa Cruz; 1:500), XBP1 (M-186; Santa Cruz;
1:500), Ubiquitin (P4D1; Covance; 1:1,000) and b-tubulin (Sigma; 1:10,000). All
primary antibodies were incubated overnight at 4 °C. Horseradish peroxidase or
Alexafluor-conjugated goat anti-rabbit (Rockland) or goat anti-mouse (Rockland)
antibodies were used as secondary antibodies (1:2,000). Western blots were
developed using an ECL detection kit (Thermo Scientific) or an Odyssey Imager
(LI-COR). Full scans of uncropped blots can be found in Supplementary Figs 9,10.
Gene expression analysis and quantitative PCR.Total RNA was isolated and
purified using the RNeasy kit (Qiagen). cDNA was obtained by reverse transcribing
1–2 mg of total RNA using SuperscriptIII Reverse Transcriptase (Invitrogen) and
used for qPCR. qPCR reactions were performed in triplicate using SYBR Green
reagents (Quanta Biosciences) on a StepOnePlus (Life Technologies). GAPDH was
used as an endogenous control. All results were normalized to GAPDH. Primer sets
used are: GAPDH: 50-AAGGTCGGAGTCAACGGATTT G-30and 50-CCATGGG
TGGAATCATATTGGAA-30; SCCA1: 50-AGCCGCGGTCTCGTGC-30and
50-GGCAGCTGCAGCTTCTG-30; SCCA2: 50-AGCCACGGTCTCTCAG-30and
5-GCAGCTGCAGCTTCCA-30; Serpinb3a: 50-CATTTGTTTGCTGAAGCCAC
TAC-30and 50-CATGTTCGAAATCCAGTGATTCC-30; Serpinb3b: 50-ATTCGT
TTTCATGCAGCTGATGT-30and 50-GAAAGCTGAAGTTAAATTTGTTCG-30;
PEA3: 50-GGACTTCGCCTACGACTCAG-30and 50-CGCAGAGGTTTCTCA
TAGCC-30; IL-6: 50-TCCACAAGCGCCTTCGGTCCA30and 50-AGGGCTGA
GATGCCGTCGAGGA-30; IL-8: 50-AAGGAAAACTGGGTGCAGAG-30and
50-ATTGCATCTGGCAACCCTAC-30; CXCL1: 50-CACCCCAAGAACATCCA
AAG-30and 50-TAACTATGGGGGATGCAGGA-30; G-CSF: 50-ACTACAAGCA
GCACTGCCCT-30and 50-AGCAGTCAAAGGGGATGACA-30; GM-CSF: 50-CAA
GTGAGGAAGATCCAGGG-30; and 50-AGAGAGTGTCCGAGCAGCAC-30.
Cytokine array.Cells were washed once with PBS and incubated for 8 h with fresh
media. Supernatant was collected, cleared by centrifugation and used immediately.
The amount of supernatant used was normalized to cell number and used with the
human cytokine array kit (R&D Systems). IRDye 800CW Streptavidin (Rockland)
was used as secondary (1:2,000) and arrays were imaged and quantified on an
Odyssey Imager.
Senescence assays.Senescence was induced by oncogenic Ras, etoposide
(100 mM, 48 h), H
2
O
2
(100 mM, 1 h) or long-term passaging (replicative senes-
cence). All cells were analysed 7 days post-selection or post-treatment. For
SA-b-gal staining, cells were fixed in 2% formaldehyde, 0.2% glutaraldehyde in PBS
for 15 min and stained (150 mM NaCl, 2 mM MgCl
2
,5mMK
3
Fe(CN)
6
,5mM
K
4
Fe(CN)
6
, 40 mM NaPi, pH 6.0, 1 mgml1X-Gal) overnight at 37 °C. For BrdU
staining, cells were cultured with BrdU (10 nM) for 6 h, fixed with acid ethanol
(90% ethanol, 5% acetic acid, 5% H
2
O) for 30 min at room temperature. Cells were
then washed once with PBS, incubated with 2 M HCl for 20 min, 0.1 M sodium
borate, pH 8.5 for 2 min and washed once with PBS. Cells were blocked in 10%
bovine serum albumin (BSA) in PBS for 1 h at room temperature and incubated
with anti-BrdU (BD Pharmingen, 1:500 in 5% BSA in 0.1% PBS-tween) overnight
at 4 °C. Cells were washed, incubated with anti-mouse Alexa-594 (1:500) for 1 h at
room temperature, washed, co-stained with 40,6-diamidino-2-phenylindole (DAPI)
and mounted.
NF-jB luciferase activity assay.NF-kB activity was determined by using an
NF-kB luciferase reporter construct where the luciferase gene is under control of
the IL-6 promoter and internal control plasmid pCMV-RL using a dual-luciferase
reporter system (Promega). Cells were plated 24 h before transfection at 5 104
cells per well of 24-well plate. NF-kB-luciferase vector (250 ng) and 100 ng
pCMV-RL were used for transfection. Twenty-four hours post transfection, cells
were washed with PBS and lysed in 100 ml passive lysis buffer for 10 min. Luciferase
activity was determined following the manufacturer’s recommended protocol with
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729
10 NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications
&2014 Macmillan Publishers Limited. All rights reserved.
a SpectraMax M5 Microplate Reader. The ratios of firefly luciferase versus renilla
luciferase are used as relative luciferase activities.
TCGA analysis.TCGA human colorectal cancer (study abbreviation:
COADREAD) data were downloaded from Broad Institute’s Genome Data
Analysis Center (GDAC). Standard data of somatic mutations (Mutation_
Packager_Calls_Level_3) and RNA expression (Merge_transcriptome__
agilentg4502a_07_3__unc_edu__Level_3__unc_lowess_normalization_gene_
level__data.Level_3) were used. There were 207 human colorectal tumours that
have both somatic mutation and mRNA expression data available. KRas was
mutated in 87 out of the 207 samples. SCCA mRNA expression level was compared
between the groups with wild-type and mutant KRas.
Immunohistochemistry.Tissue microarrays came from the Vanderbilt GI SPORE
Tissue Core. Distribution and the use of all human samples were approved by the
Institutional Review Boards of Vanderbilt University Medical Center and Stony
Brook University, and samples were obtained with informed consent. IHC was
performed on a Ventana XT (Tucson, AZ, USA) autostainer, according to the
manufacturer’s directions.
Tissue microarray analysis.Damaged core spots and those that did not contain
cancerous tissue were eliminated from scoring. The sections were scored inde-
pendently by two evaluators blinded to the clinical status of the patients. Samples
were scored as previously described10. In brief, samples were scored as percent of
tumour cells with SCCA or IL-6 expression: 0, no expression; 1, o10%; 2, 10–50%;
3, 450%. For SCCA, a score Z1 was considered positive. For IL-6, 0, negative; 1,
weak; 2, moderate; 3, strong.
Xenograft tumour experiments.Male athymic nude mice, 6- to 8 weeks old, were
obtained from Taconic Farms. Mice were housed and monitored at the Division of
Laboratory Animal Resources at Stony Brook University. All experimental proce-
dures and protocols were approved by the Stony Brook University institutional
animal care and use committee (IACUC). Tumours were established by
resuspending 1 106tumour cells in 100 ml PBS and injecting the cells into the
mid-flanks of mice using a 26-gauge needle. For each tumour, the tumour length (l)
and width (w) was measured every 4–5 days with an electronic caliper. Tumour
volume (v) was calculated using the formula v¼(lw2)/2 and plotted in mm3.
Statistical analysis.Statistical analyses were performed with GraphPad Prism
(Graphpad Software Inc). For gene expression, luciferase activity and xenograft
tumour growth significance was calculated with t-tests. w2-tests were used to
assess statistical significance of various categorical clinical features between
SCCA-negative and SCCA-positive samples. For the TCGA colorectal cancer data
analysis, Wilcoxon Rank Sum test was performed. For all tests, P-values were
considered statistically significant when o0.05.
References
1. Bos, J. L. Ras oncogenes in human cancer: a review. Cancer Res. 49, 4682–4689
(1989).
2. Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat. Rev.
Cancer 3, 11–22 (2003).
3. Sparmann, A. & Bar-Sagi, D. Ras-induced interleukin-8 expression plays a
critical role in tumor growth and angiogenesis. Cancer Cell 6, 447–458 (2004).
4. Pylayeva-Gupta, Y., Lee, K. E., Hajdu, C. H., Miller, G. & Bar-Sagi, D.
Oncogenic Kras-induced GM-CSF production promotes the development of
pancreatic neoplasia. Cancer Cell 21, 836–847 (2012).
5. Kuilman, T. et al. Oncogene-induced senescence relayed by an interleukin-
dependent inflammatory network. Cell 133, 1019–1031 (2008).
6. Coppe, J. P. et al. Senescence-associated secretory phenotypes reveal cell-
nonautonomous functions of oncogenic RAS and the p53 tumor suppressor.
PLoS Biol. 6, 2853–2868 (2008).
7. Ancrile, B., Lim, K. H. & Counter, C. M. Oncogenic Ras-induced secretion of
IL6 is required for tumorigenesis. Genes Dev. 21, 1714–1719 (2007).
8. Leslie, K. et al. Differential interleukin-6/Stat3 signaling as a function of cellular
context mediates Ras-induced transformation. Breast Cancer Res. 12, R80
(2010).
9. Schneider, S. S. et al. A serine proteinase inhibitor locus at 18q21.3 contains
a tandem duplication of the human squamous cell carcinoma antigen gene.
Proc. Natl Acad. Sci. USA 92, 3147–3151 (1995).
10. Catanzaro, J. M. et al. Elevated expression of squamous cell carcinoma antigen
(SCCA) is associated with human breast carcinoma. PLoS ONE 6, e19096
(2011).
11. Guido, M. et al. Squamous cell carcinoma antigen in human liver
carcinogenesis. J. Clin. Pathol. 61, 445–447 (2008).
12. Vassilakopoulos, T. et al. Diagnostic and prognostic significance of squamous
cell carcinoma antigen in non-small cell lung cancer. Lung Cancer 32, 137–144
(2001).
13. Cataltepe, S. et al. Co-expression of the squamous cell carcinoma antigens 1
and 2 in normal adult human tissues and squamous cell carcinomas.
J. Histochem. Cytochem. 48, 113–122 (2000).
14. McGettrick, A. F., Barnes, R. C. & Worrall, D. M. SCCA2 inhibits
TNF-mediated apoptosis in transfected HeLa cells. The reactive centre loop
sequence is essential for this function and TNF-induced cathepsin G is a
candidate target. Eur. J. Biochem. 268, 5868–5875 (2001).
15. Suminami, Y. et al. Inhibition of apoptosis in human tumour cells by the
tumour-associated serpin, SCC antigen-1. Br. J. Cancer 82, 981–989 (2000).
16. Brioschi, P. A., Bischof, P., Delafosse, C. & Krauer, F. Squamous-cell carcinoma
antigen (SCC-A) values related to clinical outcome of pre-invasive and invasive
cervical carcinoma. Int. J. Cancer 47, 376–379 (1991).
17. Scambia, G. et al. The value of squamous cell carcinoma antigen in patients
with locally advanced cervical cancer undergoing neoadjuvant chemotherapy.
Am. J. Obstet. Gynecol. 164, 631–636 (1991).
18. Ngan, H. Y., Chan, S. Y., Wong, L. C., Choy, D. T. & Ma, H. K. Serum
squamous cell carcinoma antigen in the monitoring of radiotherapy treatment
response in carcinoma of the cervix. Gynecol. Oncol. 37, 260–263 (1990).
19. Suminami, Y. et al. Promoter analyses of SCC antigen genes. Biochim. Biophys.
Acta 1727, 208–212 (2005).
20. Young, A. R. et al. Autophagy mediates the mitotic senescence transition. Genes
Dev. 23, 798–803 (2009).
21. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W. Oncogenic
ras provokes premature cell senescence associated with accumulation of p53
and p16INK4a. Cell 88, 593–602 (1997).
22. Kikuchi, H., Pino, M. S., Zeng, M., Shirasawa, S. & Chung, D. C. Oncogenic
KRAS and BRAF differentially regulate hypoxia-inducible factor-1alpha
and -2alpha in colon cancer. Cancer Res. 69, 8499–8506 (2009).
23. Baines, A. T., Xu, D. & Der, C. J. Inhibition of Ras for cancer treatment: the
search continues. Future Med. Chem. 3, 1787–1808 (2011).
24. Guo, B. & Sharrocks, A. D. Extracellular signal-regulated kinase mitogen-
activated protein kinase signaling initiates a dynamic interplay between
sumoylation and ubiquitination to regulate the activity of the transcriptional
activator PEA3. Mol. Cell Biol. 29, 3204–3218 (2009).
25. Iwasaki, M. et al. E1AF/PEA3 reduces the invasiveness of SiHa cervical cancer
cells by activating serine proteinase inhibitor squamous cell carcinoma antigen.
Exp. Cell Res. 299, 525–532 (2004).
26. Chien, Y. et al. Control of the senescence-associated secretory phenotype by
NF-kappaB promotes senescence and enhances chemosensitivity. Genes Dev.
25, 2125–2136 (2011).
27. Rodier, F. et al. Persistent DNA damage signalling triggers senescence-
associated inflammatory cytokine secretion. Nat. Cell Biol. 11, 973–979 (2009).
28. Garg, A. D. et al. ER stress-induced inflammation: does it aid or impede disease
progression? Trends. Mol. Med. 18, 589–598 (2012).
29. Ullman, E., Pan, J. A. & Zong, W. X. Squamous cell carcinoma antigen 1
promotes caspase-8-mediated apoptosis in response to endoplasmic reticulum
stress while inhibiting necrosis induced by lysosomal injury. Mol. Cell. Biol. 31,
2902–2919 (2011).
30. Denoyelle, C. et al. Anti-oncogenic role of the endoplasmic reticulum
differentially activated by mutations in the MAPK pathway. Nat. Cell Biol. 8,
1053–1063 (2006).
31. Maitra, A. & Hruban, R. H. Pancreatic cancer. Annu. Rev. Pathol. 3, 157–188
(2008).
32. Pei, H. et al. FKBP51 affects cancer cell response to chemotherapy by negatively
regulating Akt. Cancer Cell 16, 259–266 (2009).
33. Badea, L., Herlea, V., Dima, S. O., Dumitrascu, T. & Popescu, I. Combined gene
expression analysis of whole-tissue and microdissected pancreatic ductal
adenocarcinoma identifies genes specifically overexpressed in tumor epithelia.
Hepatogastroenterology 55, 2016–2027 (2008).
34. Iacobuzio-Donahue, C. A. et al. Exploration of global gene expression patterns
in pancreatic adenocarcinoma using cDNA microarrays. Am. J. Pathol. 162,
1151–1162 (2003).
35. Segara, D. et al. Expression of HOXB2, a retinoic acid signaling target in
pancreatic cancer and pancreatic intraepithelial neoplasia. Clin. Cancer Res. 11,
3587–3596 (2005).
36. Grutzmann, R. et al. Gene expression profiling of microdissected pancreatic
ductal carcinomas using high-density DNA microarrays. Neoplasia 6, 611–622
(2004).
37. Logsdon, C. D. et al. Molecular profiling of pancreatic adenocarcinoma and
chronic pancreatitis identifies multiple genes differentially regulated in
pancreatic cancer. Cancer Res. 63, 2649–2657 (2003).
38. Jackson, E. L. et al. Analysis of lung tumor initiation and progression using
conditional expression of oncogenic K-ras. Genes Dev. 15, 3243–3248 (2001).
39. Luke, C. J. et al. An intracellular serpin regulates necrosis by inhibiting the
induction and sequelae of lysosomal injury. Cell 130, 1108–1119 (2007).
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729 ARTICLE
NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications 11
&2014 Macmillan Publishers Limited. All rights reserved.
40. Wang, S. & Kaufman, R. J. The impact of the unfolded protein response on
human disease. J. Cell Biol. 197, 857–867 (2012).
41. Luo, B. & Lee, A. S. The critical roles of endoplasmic reticulum chaperones and
unfolded protein response in tumorigenesis and anticancer therapies. Oncogene
32, 805–818 (2013).
42. De Raedt, T. et al. Exploiting cancer cell vulnerabilities to develop a
combination therapy for ras-driven tumors. Cancer Cell 20, 400–413 (2011).
43. Terzic, J., Grivennikov, S., Karin, E. & Karin, M. Inflammation and colon
cancer. Gastroenterology 138, 2101–2114 e2105 (2010).
44. Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860–867
(2002).
45. Meylan, E. et al. Requirement for NF-kappaB signalling in a mouse model of
lung adenocarcinoma. Nature 462, 104–107 (2009).
46. Katagiri, C., Nakanishi, J., Kadoya, K. & Hibino, T. Serpin squamous cell
carcinoma antigen inhibits UV-induced apoptosis via suppression of c-JUN
NH2-terminal kinase. J. Cell Biol. 172, 983–990 (2006).
47. Vidalino, L. et al. SERPINB3, apoptosis and autoimmunity. Autoimmun. Rev. 9,
108–112 (2009).
48. Murakami, A. et al. Squamous cell carcinoma antigen suppresses radiation-
induced cell death. Br. J. Cancer 84, 851–858 (2001).
49. Suminami, Y. et al. Suppression of a squamous cell carcinoma (SCC)-related
serpin, SCC antigen, inhibits tumor growth with increased intratumor
infiltration of natural killer cells. Cancer Res. 61, 1776–1780 (2001).
50. Baron, J. A. et al. A randomized trial of aspirin to prevent colorectal adenomas.
New Engl. J. Med. 348, 891–899 (2003).
51. Sandler, R. S. et al. A randomized trial of aspirin to prevent colorectal
adenomas in patients with previous colorectal cancer. New Engl. J. Med. 348,
883–890 (2003).
52. Khosravi-Far, R. et al. Oncogenic Ras activation of Raf/mitogen-activated
protein kinase-independent pathways is sufficient to cause tumorigenic
transformation. Mol. Cell Biol. 16, 3923–3933 (1996).
53. Slack, J. K., Catling, A. D., Eblen, S. T., Weber, M. J. & Parsons, J. T.
c-Raf-mediated inhibition of epidermal growth factor-stimulated cell
migration. J. Biol. Chem. 274, 27177–27184 (1999).
Acknowledgements
We thank Drs Alea Mills, Scott Lowe, Masashi Narita, Scott Eblen, Andrew Sharrocks
and Erich Mackow for reagents and Drs Richard Lin and Dafna Bar-Sagi for critical
reading. J.M.C. was supported by the NCI T32 training grant (T32CA009176). This work
was supported by grants from NIH (R01CA129536 and R01GM97355 to W.-X.Z.,
U01CA168409 to R.S.P., and R01CA159222 and R01CA100126 to H.C.C.), and the Carol
Baldwin Breast Cancer Research Foundation (to W.-X.Z.). P50CA095103 for the
Vanderbilt University Medical Center GI Spore Tissue Core.
Author contributions
J.M.C., N.S., J.-A.P., Y.S. and C.S. performed experiments. J.L. R.S.P., and H.C.C. aided
with data analysis. J.M.C. and W.-X.Z. wrote the paper.
Additional information
Supplementary Information accompanies this paper at http://www.nature.com/
naturecommunications
Competing financial interests: The authors declare no competing financial interests.
Reprints and permission information is available online at http://npg.nature.com/
reprintsandpermissions/
How to cite this article: Catanzaro, J. M. et al. Oncogenic Ras induces inflammatory
cytokine production by upregulating the squamous cell carcinoma antigens SerpinB3/B4.
Nat. Commun. 5:3729 doi: 10.1038/ncomms4729 (2014).
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms4729
12 NATURE COMMUNICATIONS | 5:3729 | DOI: 10.1038/ncomms4729 | www.nature.com/naturecommunications
&2014 Macmillan Publishers Limited. All rights reserved.
... SERPINB3 mediates suppressive chemokine production through the promotion of STAT activation. Although SERPINB3 has been implicated in proinflammatory signaling in pancreatic cancer and Kras-mutant tumors (26), the underlying molecular mechanism is unknown. To provide further insight into the SERPINB3-mediated suppressive immune response, we used a human phosphorylation pathway profiling array that contained 5 cancer-associated pathways -MAPK, AKT, JAK/STAT, NF-κB, and TGF-β -and identified 14 proteins with upregulated phosphorylation (fold change ≥2) and 4 proteins with downregulated phosphorylation (fold change ≤ 0.5) in Caski/B3 versus Caski/Ctrl cells (Supplemental Figure 9). ...
... The association between SERPINB3 and chemokines has been reported in atopic dermatitis and psoriasis, in which downregulation of SERPINB3 in keratinocytes was associated with reduced expression of CXCL1/-5/-8 (27) and S100A8 (16). Catanzaro and colleagues showed that SERPINB3 is a downstream mediator of mutant Ras-induced tumorigenesis and that KD of SERPINB3 led to decreased production of IL-6, CXCL1, and CXCL8, thereby suppressing tumorigenesis (26). In patients with cervical cancer or esophageal squamous cell carcinoma, high expression of SERPINB3 was associated with lymph node metastasis (28)(29)(30); however, the underlying causes are unknown. ...
Article
Full-text available
Cancer patients with high serum squamous cell carcinoma antigen (SCCA1/SERPINB3) are commonly associated with treatment resistance and poor prognosis. Despite being a clinical biomarker, the modulation of SERPINB3 in tumor immunity is poorly understood. We found positive correlations of SERPINB3 with CXCL1/8, S100A8/A9 and myeloid cell infiltration through RNAseq analysis of human primary cervix tumors. Induction of SERPINB3 resulted in increased CXCL1/8 and S100A8/A9, which promoted monocyte and MDSC migration in vitro. In mouse models, Serpinb3a-tumors showed increased MDSC and TAM infiltration contributing to T cell inhibition and this was further augmented upon radiation. Intratumoral knockdown of Serpinb3a demonstrated tumor growth inhibition and reduced CXCL1, S100A8/A9, MDSC, and M2 macrophage infiltration. These changes led to enhanced cytotoxic T cell function and sensitized tumors to radiotherapy. We further revealed SERPINB3 promoted STAT-dependent suppressive chemokine expression, whereby inhibiting STAT activation by ruxolitinib or siRNA abrogated CXCL1/8 and S100A8/A9 in SERPINB3 cells. Patients with elevated pre-treatment SCCA and high pSTAT3 had increased intratumoral CD11b+ myeloid cell compared to patients with low SCCA and pSTAT3 cohort that had overall improved survival after radiotherapy. These findings provide a preclinical rationale for targeting SERPINB3 in tumors to counteract the immunosuppression and improve response to radiation.
... The interaction between SAA and its formyl peptide receptor 2 can activate NF-κB, inducing IL-8 production [66]. SCCA inhibits protein turnover and induces an unfolded protein response, which in turn activates NF-κB, promoting the expression of IL-6 [67]. HGF enhances IL-6 secretion by stimulating the mTOR/IL1A/NF-κB signaling pathway [68].The sIL2Rα, which is a downstream target of NF-κB [69], shows a significant positive correlation with the severity of CC. ...
Article
Full-text available
The nuclear factor-κB (NF-κB) family, consisting of several transcription factors, has been implicated in the regulation of cell proliferation and invasion, as well as inflammatory reactions and tumor development. Cervical cancer (CC) results from long-term interactions of multiple factors, among which persistent high-risk human papillomavirus (hrHPV) infection is necessary. During different stages from early to late after HPV infection, the activity of NF-κB varies and plays various roles in carcinogenesis and progress of CC. As the center of the cell signaling transduction network, NF-κB can be activated through classical and non-classical pathways, and regulate the expression of downstream target genes involved in regulating the tumor microenvironment and acquiring hallmark traits of CC cells. Targeting NF-κB may help treat CC and overcome the resistance to radiation and chemotherapy. Even though NF-κB inhibitors have not been applied in clinical treatment as yet, due to limitations such as dose-restrictive toxicity and poor tumor-specificity, it is still considered to have significant therapeutic potential and application prospects. In this review, we focus on the role of NF-κB in the process of CC occurrence and hallmark capabilities acquisition. Finally, we summarize relevant NF-κB-targeted treatments, providing ideas for the prevention and treatment of CC.
... Previous studies have established that NF-κB is activated in a RAS-dependent activation pathway through IL-1 binding to AcP (IL-1 accessory protein) [26] and its co-receptor TILRR [15]. In cardiac fibroblasts, IL-1β-mediated activation of NF-κB contributes to IL-1β-induced IL-8 mRNA expression [24]. ...
Article
Full-text available
Glycosaminoglycans (GAGs) are long linear polysaccharides found in every mammalian tissue. Previously thought only to be involved in cellular structure or hydration, GAGs are now known to be involved in cell signaling and protein modulation in cellular adhesion, growth, proliferation, and anti-coagulation. In this study, we showed that GAGs have an inhibitory effect on the IL-1β-stimulated mRNA expression of IL-6 and IL-8. Exogenous heparin (p < 0.0001), heparan (p < 0.0001), chondroitin (p < 0.049), dermatan (p < 0.0027), and hyaluronan (p < 0.0005) significantly reduced the IL-1β-induced IL-8 mRNA expression in HeLa cells. Exogenous heparin (p < 0.0001), heparan (p < 0.0001), and dermatan (p < 0.0027) also significantly reduced IL-1β-induced IL-6 mRNA expression in HeLa cells, but exogenous chondroitin and hyaluronan had no significant effect. The exogenous GAGs may reduce the transcription of these inflammatory cytokines through binding to TILRR, a co-receptor of IL-1R1, and block/reduce the interactions of TILRR with IL-1R1.
... Liver tumors with stemness signatures are highly aggressive, and in hepatocellular carcinoma (HCC), the subset of more aggressive tumors, characterized by early recurrence after surgical resection, have shown the highest levels of SerpinB3, associated with high βcatenin and TGF-β1 expression [32]. Additionally, in vitro data have shown that SerpinB3 promotes the EMT and increases cell proliferation and invasiveness [33], while oncogenic Ras upregulates SerpinB3/4 expression, leading to NF-kB activation, IL-6 production and tumor growth [34,35]. SerpinB3/4 isoforms are identifiable in serum through binding to IgM complexes and as free-circulating proteins [36]. ...
Article
Full-text available
Simple Summary Cholangiocarcinoma is characterized by a very poor outcome and SerpinB3, a serine protease inhibitor, has recently been found to play a relevant role in malignant transformation in different cancers. The aim of this study was to analyze the expression of this biomarker in the serum and surgical specimens of cholangiocarcinoma in relation to clinical outcome. High levels of SerpinB3/4 were detected in tumoral tissue in 12.2% of CCA, which were characterized by a more advanced TNM stage and lower overall patient survival, independently of CCA subclass. In addition, patients who had detectable free or IgM-linked SerpinB3/4 in serum showed poorer survival. In conclusion, the present study provides evidence that SerpinB3/4, both in the serum and in tumoral tissue, could be considered a useful biomarker to identify the small subgroup of CCA patients with more aggressive tumor biology and dismal prognosis. Abstract Cholangiocarcinoma (CCA), the second most common primary liver tumor, is associated with a dismal outcome, and useful prognostic markers are not currently available in clinical practice. SerpinB3, a serine protease inhibitor, was recently found to play a relevant role in malignant transformation in different cancers. The aim of the present study was to determine the expression of SerpinB3/4 in tissue and serum samples of patients with CCA in relation to clinical outcomes. SerpinB3/4 was assessed in the tissue microarrays (TMAs) of 123 surgically resected CCAs. ELISA assays were carried out in 188 patients with CCA to detect the free and IgM-linked forms of SerpinB3/4. Overall survival was analyzed in relation to SerpinB3/4 expression, and Cox models were used to identify the variables associated with survival. High levels of SerpinB3/4 (TMA score 2+/3+) were detected in 15 tumors (12.2%), characterized by a more advanced TNM stage (III/IV: 64.3% vs. 31.3%; p = 0.031) and lower overall patient survival, independently of CCA subclass (intrahepatic CCA: median 1.1 (0.8—Not Estimable, NE) vs. 2.4 (1.8–3.4) years; p = 0.0007; extrahepatic CCA: median 0.8 (0.2—NE) vs. 2.2 (1.5–5.4) years; p = 0.011). Vascular invasion (p = 0.027) and SerpinB3/4 scores (p = 0.0016) were independently associated with mortality in multivariate analysis. Patients who had detectable free or IgM-linked SerpinB3/4 in their serum showed poorer survival (1 vs. 2.4 years, p = 0.015, for free SerpinB3/4, and 1 vs. 2.6 years, p = 0.0026, for SerpinB3/4–IgM). In conclusion, high levels of SerpinB3/4 in tissue and serum in CCA are associated with poor outcomes after surgery, regardless of tumor subclass.
... [18][19][20] SerpinB3 is indeed up-regulated in the liver by oxidative stress, iron overload, hypoxia and chronic inflammation. [21][22][23] Recent findings have characterised SerpinB3 as a novel pro-inflammatory mediator in non-alcoholic steatohepatitis (NASH), as this molecule, produced and released by damaged or stressed hepatocytes, plays an important role among the signals that contribute to promoting the pro-inflammatory phenotype of liver macrophages in NASH. 24 In addition, it has also been demonstrated that SerpinB3 is able to directly activate human HSC, resulting in a strong up-regulation of genes involved in fibrogenesis and angiogenesis. ...
Article
Full-text available
Background SerpinB3 is a cysteine protease inhibitor involved in liver disease progression due to its proinflammatory and profibrogenic properties. The polymorphic variant SerpinB3‐PD (SB3‐PD), presents a substitution in its reactive centre loop, determining the gain of function. Aims To disclose the clinical characteristics of a cohort of patients with cirrhosis in relation to the presence of SB3‐PD and to assess the effect of this genetic variant on fibrogenic and inflammatory cytokines in vitro. Methods We assessed SB3 polymorphism in 90 patients with cirrhosis, prospectively followed up in our referral centre. We used HepG2 and HuH‐7 cells transfected to overexpress either wild‐type SB3 (SB3‐WT) or SB3‐PD to assess their endogenous effect, while LX2 and THP‐1 cells were treated with exogenous SB3‐WT or SB3‐PD proteins. Results Patients carrying SB3‐PD had more severe portal hypertension and higher MELD scores, than patients carrying SB3‐WT. In multivariate analysis, SB3‐PD was an independent predictor of cirrhosis complications. Patients with SB3‐PD polymorphism presented with more severe liver fibrosis and inflammatory features. Hepatoma cells overexpressing SB3‐PD showed higher TGF‐β1 expression than controls. The addition of recombinant SB3‐PD induced an up‐regulation of TGF‐β1 in LX2 cells and a more prominent inflammatory profile in THP‐1 cells, compared to the effect of SB3‐WT protein. Conclusions The polymorphic variant SB3‐PD is highly effective in determining activation of TGF‐β1 and inflammation in vitro. Patients with cirrhosis who carry SB3‐PD polymorphism may be more prone to develop severe liver disease progression. However, further validation studies are warranted to support the in vivo relevance of this polymorphism.
... It is well known that SB3/4 is localized predominantly in the cytosol; however, it has been also detected in other subcellular compartments, including the nucleus, both in cells irradiated with UV rays [38] and in the liver of mice transgenic for SerpinB3 [42,43]. In addition, in clinical samples, nuclear SB3/4 has been reported in various cancers [44][45][46], besides cytosolic localization. Further studies are needed to assess the clinical or prognostic value of specific SB3 detection at the nuclear level. ...
Article
Full-text available
SerpinB3 is a serine protease inhibitor that plays a relevant role in disease progression and cancer by increasing fibrosis, cell proliferation, and invasion, besides conferring resistance to apoptosis. The mechanisms underlying these biological activities are not yet fully understood. The aim of this study was to generate antibodies directed against different SerpinB3 epitopes to better investigate their biological role. Five exposed epitopes were identified using the software DNASTAR Lasergene and the corresponding synthetic peptides were used for NZW rabbit immunization. Anti-P#2 and anti-P#4 antibodies were able to recognize both SerpinB3 and SerpinB4 by ELISA. Anti-P#5 antibody, produced against the reactive site loop of SerpinB3, showed the greatest specific reactivity for human SerpinB3. This antibody was able to recognize SerpinB3 at nuclear level, while anti-P#3 antibody recognized SerpinB3 only at cytoplasmic level, both by immunofluorescence and by immunohistochemistry. The biological activity of each antibody preparation was assessed in HepG2 cells overexpressing SerpinB3 and anti-P#5 antibody reduced proliferation by 12% cell and cell invasion by 75%, while trivial results were obtained with the other antibody preparations. These findings indicate that the reactive site loop of SerpinB3 is essential for the invasiveness features induced by this serpin and it could become a novel druggable target.
... This included a robust expression of the SERPINB family of serine protease inhibitors, which are clustered at chr.18q21 (12,13). SERPINB4 expression has previously been described as regulated downstream of oncogenes such as Ras (44). Our findings here suggest that STAT3, which is a major tyrosine phosphorylation target of the various NSCLC ALK fusions (EML4-ALK-V1, EML4-ALK-V3, TFG-ALK, and KIF5B-ALK), is also important for the expression of SERPINB4. ...
Article
Anaplastic lymphoma kinase (ALK) fusion variants in non-small-cell-lung cancer (NSCLC) consist of numerous dimerising fusion partners, with the most common being EML4. Clinical data suggests that the degree of treatment benefit in response to ALK tyrosine kinase inhibitors (TKIs) differs among the variant present in the patient tumor. Therefore, a better understanding the oncogenic signaling networks driven by different ALK-fusion variants is important. Here, we developed highly controlled doxycycline-inducible cell models bearing four different ALK fusion proteins, namely EML4-ALK-V1, EML4-ALK-V3, KIF5B-ALK, and TFG-ALK, in the context of non-tumorigenic NL20 human bronchial epithelial cells. These were complimented by patient-derived NSCLC cell lines harboring either EML4-ALK-V1 or EML4-ALK-V3 fusions. RNA-seq and phosphoproteomics analysis were employed to identify dysregulated genes and hyper/hypo-phosphorylated proteins associated with ALK fusion expression. Among ALK fusion induced responses, we noted a robust inflammatory signature that included up-regulation of the Serpin B4 serine protease inhibitor in both NL20-inducible cell models and ALK-positive NSCLC patient-derived cell lines. We show that STAT3 is a major transcriptional regulator of SERPINB4 downstream of ALK fusions, along with NF-kB and AP1. The upregulation of SERPINB4 promotes survival of ALK fusion expressing cells and inhibits natural killer (NK) cell-mediated cytotoxicity. In conclusion, our study reveals a novel ALK downstream survival axis that regulates Serpin B4 expression and identifies a molecular target that has potential for therapeutic impact targeting the immune response together with ALK TKIs in NSCLC. Citation Format: Tzu-Po Chuang, Wei-Yun Lai, Jonatan L. Gabre, Dan E. Lind, Ganesh Umapathy, Abdulmalik A. Bokhari, Bengt Bergman, Linnea Kristenson, Fredrik B. Thorén, Anh Le, Robert Doebele, Jimmy V. Eynden, Ruth H. Palmer, Bengt Hallberg. ALK fusion oncogene driven SERPINB4 expression enhances tumor survival in NSCLC. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3936.
Article
A normal somatic cell undergoes cycles of finite cellular divisions. The presence of surveillance checkpoints arrests cell division in response to stress inducers: oxidative stress from excess free radicals, oncogene-induced abnormalities, genotoxic stress, and telomere attrition. When facing such stress when undergoing these damages, there is a brief pause in the cell cycle to enable repair mechanisms. Also, the nature of stress determines whether the cell goes for repair or permanent arrest. As the cells experience transient or permanent stress, they subsequently choose the quiescence or senescence stage, respectively. Quiescence is an essential stage that allows the arrested/damaged cells to go through appropriate repair mechanisms and then revert to the mainstream cell cycle. However, senescent cells are irreversible and accumulate with age, resulting in inflammation and various age-related disorders. In this review, we focus on senescence-associated pathways and therapeutics understanding cellular senescence as a cascade that leads to aging, while discussing the recent details on the molecular pathways involved in regulating senescence and the benefits of therapeutic strategies against accumulated senescent cells and their secretions. ARTICLE HISTORY
Article
Pancreatic ductal adenocarcinoma (PDAC) exhibits distinct molecular subtypes: classical/progenitor and basal-like/squamous. Our study aimed to identify genes contributing to the development of the basal-like/squamous subtype, known for its aggressiveness. Transcriptome analyses revealed consistent upregulation of SERPINB3 in basal-like/squamous PDAC, correlating with reduced patient survival. SERPINB3 transgene expression in PDAC cells enhanced in vitro invasion and promoted lung metastasis in a mouse PDAC xenograft model. Metabolome analyses unveiled a metabolic signature linked to both SERPINB3 and the basal-like/squamous subtype, characterized by heightened carnitine/acylcarnitine and amino acid metabolism, associated with poor prognosis in patients with PDAC and elevated cellular invasiveness. Further analysis uncovered that SERPINB3 inhibited the cysteine protease calpain, a key enzyme in the MYC degradation pathway, and drove basal-like/squamous subtype and associated metabolic reprogramming through MYC activation. Our findings indicate that the SERPINB3-MYC axis induces the basal-like/squamous subtype, proposing SERPINB3 as a potential diagnostic and therapeutic target for this variant.
Article
Psoriasis is a chronic recurrent skin disease, with excessive proliferation of keratinocytes. Recent studies indicated the pathogenic roles of circular RNA (circRNA) in psoriasis. Here, we screened the circRNA profiles from five psoriatic skin lesions and five normal skin tissues by circRNA sequencing and identified 1118 differentially expressed circRNAs (DECs) between psoriatic and normal groups. Among these DECs, high abundant circARNTL2 has been proven upregulated in psoriatic skin lesions by RT-qPCR assay. Then, the head-to-tail structure of circARNTL2 was validated by Sanger sequencing and RNase R digestion assay. Moreover, we determined cytoplastic location of circARNTL2 by RT-qPCR assay of nuclear/cytoplasmic RNA and FISH analysis. Further experiments demonstrated that silencing circARNTL2 expression could block cell proliferation and cell cycle progression of keratinocytes. Mechanistically, circARNTL2 can bind to and regulate Serpin B4 which also affects the proliferation of keratinocytes. These findings provide evidence for the role of circARNTL2 in psoriasis.
Article
Full-text available
Article
Katagiri et al. 2006. J. Cell Biol. doi:10.1083/jcb.200508064 [OpenUrl][1][Abstract/FREE Full Text][2] [1]: {openurl}?query=rft_id%253Dinfo%253Adoi%252F10.1083%252Fjcb.200508064%26rft_id%253Dinfo%253Apmid%252F16549498%26rft.genre%253Darticle%26rft_val_fmt%253Dinfo%253Aofi%252Ffmt%253Akev%253Amtx%
Article
The squamous cell carcinoma antigens, SCCA1 and SCCA2, are members of the serine protease inhibitors (serpin) superfamily and are transcribed by two tandomly arrayed genes. A number of serpins are known to inhibit apoptosis in mammalian cells. In this study we demonstrate the ability of SCCA2 to inhibit tumor necrosis factor-alpha (TNFα)-induced apoptosis. HeLa cells stably transfected with SCCA2 cDNA had increased percentage cell survival and reduced DNA fragmentation. We investigated if the reactive centre loop (RCL) was necessary to allow SCCA2 to inhibit TNFα-mediated apoptosis. The RCL amino acids (E353Q, L354G, S355A), flanking the predicted cleavage site, were mutated and the resulting SCCA2 lost both the ability to inhibit cathepsin G and to protect stably transfected cells from TNFα-induced apoptosis. The presence of SCCA2 caused a decrease in the activation of caspase-3 upon induction with TNFα but no direct inhibition of caspases by SCCA2 has been found. Expression of cathepsin G was found to be induced in HeLa cells following treatment with TNFα. This protease has recently been shown to have a role in apoptosis through cleavage of substrates, so maybe the relevant target for SCCA2 in this system.
Article
The past two decades have witnessed an explosion in Our understanding of pancreatic cancer, and it is now clear that pancreatic cancer is a disease of inherited (germ-line) and somatic gene mutations. The genes mutated in pancreatic cancer include KRAS2, p16/CDKN2A, TP53, and SMAD4/DPC4, and these are accompanied by a substantial compendium of genomic and transcriptomic alterations that facilitate cell cycle deregulation, cell survival, invasion, and metastases. Pancreatic cancers do not arise de novo, and three distinct precursor lesions have been identified. Experimental models of pancreatic cancer have been developed in genetically engineered mice, which recapitulate the multistep progression of the cognate human disease. Although the putative cell of origin for pancreatic cancer remains elusive, minor populations of cells with stem-like properties have been identified that appear responsible for tumor initiation, metastases, and resistance of Pancreatic cancer to conventional therapies.
Article
Oncogenic ras can transform most immortal rodent cells to a tumorigenic state. However, transformation of primary cells by ras requires either a cooperating oncogene or the inactivation of tumor suppressors such as p53 or p16. Here we show that expression of oncogenic ras in primary human or rodent cells results in a permanent G1 arrest. The arrest induced by ras is accompanied by accumulation of p53 and p16, and is phenotypically indistinguishable from cellular senescence. Inactivation of either p53 or p16 prevents ras-induced arrest in rodent cells, and E1A achieves a similar effect in human cells. These observations suggest that the onset of cellular senescence does not simply reflect the accumulation of cell divisions, but can be prematurely activated in response to an oncogenic stimulus. Negation of ras-induced senescence may be relevant during multistep tumorigenesis.
Article
Different lines of research have revealed that pathways activated by the endoplasmic reticulum (ER) stress response induce sterile inflammation. When activated, all three sensors of the unfolded protein response (UPR), PERK, IRE1, and ATF6, participate in upregulating inflammatory processes. ER stress in various cells plays an important role in the pathogenesis of several diseases, including obesity, type 2 diabetes, cancer, and intestinal bowel and airway diseases. Moreover, it has been suggested that ER stress-induced inflammation contributes substantially to disease progression. However, this generalization can be challenged at least in the case of cancer. In this review, we emphasize that ER stress can either aid or impede disease progression via inflammatory pathways depending on the cell type, disease stage, and type of ER stressor.