ArticlePDF Available

SCF, Regulated by HIF-1α, Promotes Pancreatic Ductal Adenocarcinoma Cell Progression

PLOS
PLOS ONE
Authors:

Abstract and Figures

Stem cell factor (SCF) and hypoxia-inducible factor-1α (HIF-1α) both have important functions in pancreatic ductal adenocarcinoma (PDAC). This study aims to analyze the expression and clinicopathological significance of SCF and HIF-1α in PDAC specimens and explore the molecular mechanism at PDAC cells in vitro and in vivo. We showed that the expression of SCF was significantly correlated with HIF-1α expression via Western blot, PCR, chromatin immunoprecipitation (ChIP) assay, and luciferase assay analysis. The SCF level was also correlated with lymph node metastasis and the pathological tumor node metastasis (pTNM) stage in PDAC samples. The SCF higher-expression group had significantly lower survival rates than the SCF lower-expression group (p<0.05). Hypoxia up-regulated the expression of SCF through the hypoxia-inducible factor (HIF)-1α in PDAC cells at the protein and RNA levels. When HIF-1α was knocked down by RNA interference, the SCF level decreased significantly. Additionally, ChIP and luciferase results demonstrated that HIF-1α can directly bind to the hypoxia response element (HRE) region of the SCF promoter and activate the SCF transcription under hypoxia. The results of colony formation, cell scratch, and transwell migration assay showed that SCF promoted the proliferation and invasion of PANC-1 cells under hypoxia. Furthermore, the down-regulated ability of cell proliferation and invasion following HIF-1α knockdown was rescued by adding exogenous SCF under hypoxia in vitro. Finally, when the HIF-1α expression was inhibited by digoxin, the tumor volume and the SCF level decreased, thereby proving the relationship between HIF-1α and SCF in vivo. In conclusion, SCF is an important factor for the growth of PDAC. In our experiments, we proved that SCF, a downstream gene of HIF-1α, can promote the development of PDAC under hypoxia. Thus, SCF might be a potential therapeutic target for PDAC.
Content may be subject to copyright.
RESEARCH ARTICLE
SCF, Regulated by HIF-1α, Promotes
Pancreatic Ductal Adenocarcinoma Cell
Progression
Chuntao Gao
, Shasha Li
, Tiansuo Zhao
, Jing Chen, He Ren, Huan Zhang,
Xiuchao Wang, Mingxiao Lang, Jingcheng Liu, Song Gao, Xiao Zhao, Jun Sheng,
Zhanna Yuan, Jihui Hao*
Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key
Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin, 300060, China
These authors contributed equally to this work.
*haojihui@tjmuch.com
Abstract
Stem cell factor (SCF) and hypoxia-inducible factor-1α(HIF-1α) both have important func-
tions in pancreatic ductal adenocarcinoma (PDAC). This study aims to analyze the expres-
sion and clinicopathological significance of SCF and HIF-1αin PDAC specimens and
explore the molecular mechanism at PDAC cells in vitro and in vivo. We showed that the ex-
pression of SCF was significantly correlated with HIF-1αexpression via Western blot, PCR,
chromatin immunoprecipitation (ChIP) assay, and luciferase assay analysis. The SCF level
was also correlated with lymph node metastasis and the pathological tumor node metasta-
sis (pTNM) stage in PDAC samples. The SCF higher-expression group had significantly
lower survival rates than the SCF lower-expression group (p<0.05). Hypoxia up-regulated
the expression of SCF through the hypoxia-inducible factor (HIF)-1αin PDAC cells at the
protein and RNA levels. When HIF-1αwas knocked down by RNA interference, the SCF
level decreased significantly. Additionally, ChIP and luciferase results demonstrated that
HIF-1αcan directly bind to the hypoxia response element (HRE) region of the SCF promoter
and activate the SCF transcription under hypoxia. The results of colony formation, cell
scratch, and transwell migration assay showed that SCF promoted the proliferation and in-
vasion of PANC-1 cells under hypoxia. Furthermore, the down-regulated ability of cell prolif-
eration and invasion following HIF-1αknockdown was rescued by adding exogenous SCF
under hypoxia in vitro. Finally, when the HIF-1αexpression was inhibited by digoxin, the
tumor volume and the SCF level decreased, thereby proving the relationship between HIF-
1αand SCF in vivo. In conclusion, SCF is an important factor for the growth of PDAC. In our
experiments, we proved that SCF, a downstream gene of HIF-1α, can promote the develop-
ment of PDAC under hypoxia. Thus, SCF might be a potential therapeutic target for PDAC.
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 1/14
OPEN ACCESS
Citation: Gao C, Li S, Zhao T, Chen J, Ren H, Zhang
H, et al. (2015) SCF, Regulated by HIF-1α, Promotes
Pancreatic Ductal Adenocarcinoma Cell Progression.
PLoS ONE 10(3): e0121338. doi:10.1371/journal.
pone.0121338
Academic Editor: Surinder K. Batra, University of
Nebraska Medical Center, UNITED STATES
Received: August 12, 2014
Accepted: January 30, 2015
Published: March 23, 2015
Copyright: © 2015 Gao et al. This is an open access
article distributed under the terms of the Creative
Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This work was supported by grants as
follows: National Natural Science Foundation of
China (grant No. 81302082, 81272685, 31301151,
81172355); Key Program of Natural Science
Foundation of Tianjin (grant No. 11JCZDJC18400,
13YCYBYC37400); Major Anticancer Technologies R
& D Program of Tianjin (grant No. 12ZCDZSY16700).
The funders had no role in study design, data
collection and analysis, decision to publish, or
preparation of the manuscript.
Introduction
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with poor prognosis.
Understanding the molecular basis of the disease is highly desirable for developing new strate-
gies to prevent and treat PDAC [1].
Stem cell factor (SCF), also known as a mast cell growth factor, steel factor, and kit ligand
[2], is a multifunctional cytokine involved in tumor progression. SCF and its receptor, c-kit li-
gand (KL), are up-regulated in particular human malignancies including gastrointestinal stro-
mal tumor (GISTs) [3], breast cancer [4,5], hematopoietic cell [6], myeloid leukaemia [7], and
glioma [8]. The binding of SCF to c-kit causes receptor dimerization and protein kinase activa-
tion and mediates a variety of biological effects in tumor by many signal transduction pathways
[9,10]. Recently, more and more studies showed that the SCF/c-kit system has an important
function in angiogenesis, proliferation, and invasion in tumor cells [11]. Moreover, the SCF/c-
kit binding has been reported to increase hypoxia-inducible factor-1α(HIF-1α) protein syn-
thesis by the PI3K and Ras/MEK/ERK pathways in pancreatic cancer cells under normoxia,
and hypoxia up-regulated SCF gene expression in breast cancer cells through HIF-1α[5].
However, the interaction between HIF-1αand SCF in pancreatic cancer remains unclear.
As an important transcription factor, HIF-1αhas important functions in cancerous trans-
formation, chemoradiotherapy resistance, and tumor progression [12]. Tumor cells increase
the expression of HIF-1αby activating AKT under normoxia [13]. HIF-1αregulates many
downstream genes, such as erythropoietin, VEGF, heme oxygenase-1, enolase, lactate dehydro-
genase A, and aldolase [14,15]. The HIF-1αexpression level was high in pancreatic cancer, and
HIF-1αwas related to clinical stage and lymph node metastasis [16]. Therefore, HIF-1αhad
been considered as a new therapeutic target for pancreatic cancer, and targeted therapy against
HIF-1 expression in PDAC was recently investigated [1719].
In the present study, we investigated the prognostic value of HIF-1αand SCF protein ex-
pression in primary PDAC tissues. The correlation between HIF-1αand SCF was explored and
verified both in vitro and in vivo. Moreover, the biological effects of SCF on PDAC were inves-
tigated in vitro.
Materials and Methods
Cell cultures and treatments
BxPC-3 and PANC-1 cell lines were chosen for this experiment. They were obtained from the
Committee of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China).
PANC-1 was maintained in Dulbecco's modified Eagle's medium (Hyclone, USA) supple-
mented with 10% fetal bovine serum (Gibco, USA). BxPC-3 cells were maintained in RPMI-
1640 medium (Hyclone, USA) supplemented with 10% fetal bovine serum (Gibco, USA). The
cells were incubated at 37°C in a humidified atmosphere of 95% air and 5% CO
2
. For the hyp-
oxia treatment, the cells were placed in a modular incubator (Thermo Electron Co, Forma,
MA) consisting of 94% N
2
,5%CO
2
and 1% O
2
.
Reagents and antibodies
For IHC analysis: mouse monoclonal SCF antibody (sc-13126, 1:100 dilution) and mouse
monoclonal HIF-1αantibody (sc-13515, 1:100 dilution) were obtained from Santa Cruz
Biotechnology.
For western blot analysis: mouse monoclonal HIF-1αantibody (sc-13515, 1:500 dilution),
mouse monoclonal β-actin antibody (sc-8432, 1:2500 dilution), and rabbit polyclonal SCF
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 2/14
Competing Interests: The authors have declared
that no competing interests exist.
antibody (sc-9132, 1:1000 dilution) were obtained from Santa Cruz Biotechnology. The sec-
ondary antibodies preparation was either anti-rabbit (1:5000) or anti-mouse (1:5000).
For cell functional experiments: rabbit polyclonal neutralizing SCF antibody (ab9716,
0.01 μg/mL) was obtained from Abcam.
Ethics statement
The use of human samples in this study was approved by the Ethics Committee of Tianjin Can-
cer Hospital. The research involving human participants and animal experiments had been ap-
proved by our hospital and our equivalent committee. The participants provided their written
informed consents to participate in this study, but these cannot be included in the report be-
cause of the large volume and the consents were written in Chinese.
All animal experiments were conducted according to relevant national and international
guidelines.
This study was carried out in strict accordance with the recommendations in the Guide for
the Care and Use of Laboratory Animals of the National Institutes of Health (Tianjin Cancer
Hospital). The protocol was approved by the Committee on the Ethics of Animal Experiments
of the Tianjin Cancer Hospital. All surgery was performed under sodium pentobarbital anes-
thesia, and all efforts were made to minimize suffering.
Immunohistochemistry (IHC)
After obtaining the approval from the Ethics Committee, PDAC tumor samples were obtained
from 95 patients (ages, 3679 y) undergoing surgical resection, with IHC diagnosis of PDAC at
the Tianjin Cancer Institute & Hospital between July 1997 and April 2010. Specimens were cut,
deparaffinized, and rehydrated with xylene and graded alcohols. Antigen retrieval was carried
out in 5 mM citrate buffer. After the inactivation of endogenous peroxidase with 3% H
2
O
2
, the
sections were blocked with goat serum and incubated with either HIF-1αantibody (1:100) or
SCF antibody (1:100) overnight at 4°C. The sections were first rinsed in phosphate buffered sa-
line (PBS), incubated with biotinylated secondary antibody at 37°C for 20 min, and then
washed with PBS three times. Diaminobenzidine was used as a chromogen substrate. Finally,
the sections were counterstained with haematoxylin. The results of IHC staining were evaluat-
ed as follows: the intensity of tissue staining was graded as low shading (1), medium shading
(2), and high shading (3). The proportion of positive cells was assessed as 1 (1%-33% cells
stained), 2 (33%-67% cells stained), and 3 (>67% cells stained). The cases were classified into
positive groups (23. low; 46. medium; >6. high) by the product of the intensity and propor-
tion of the immunostained cancer cells for HIF-1αor SCF. Two independent pathologists
evaluated the slides, and all cases with discrepant interpretations were discussed using a dou-
ble-headed microscope until a consensus was reached. The clinicopathologic data and patient
outcomes were not given to both pathologists.
Western blotting
Total cell extracts were lysed using a RIPA lysis buffer (Beyotime, China) supplemented with
proteinase inhibitors cocktail (Sigma, USA). The protein concentration was measured using
the BCA assay kit (Sigma, USA). First, 20 μg of protein from total cell lysates was separated by
10% SDSpolyacrylamide gel electrophoresis then transferred to PVDF membranes (Invitro-
gen, USA). The membranes were then incubated in a blocking buffer containing 5% nonfat dry
milk for 1 hr at room temperature and were incubated with the different primary antibodies
overnight at 4°C. The membrane samples were then washed with TBS-T 3 times and incubated
with the corresponding secondary antibodies for 1 hr at room temperature. We used ECL
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 3/14
western blotting substrate (Pierce, USA) to test the immunoblotted bands. The primary anti-
body preparations were as follows: HIF-1αantibody, SCF antibody, and β-actin antibody.
Real-time PCR analysis
Total RNA was extracted from the two pancreatic cancer cell lines by using a TRIzol reagent
(Invitrogen, USA). Total RNA aliquots (1μg) were reverse transcribed with oligo (dT) primers
at 70°C for 10 mins. They were then placed on ice for 2 min, and they were added to the 8U Re-
verse Transcriptase reaction liquid (TaKaRa, China). The mixture was then incubated at 42°C
for 60 mins then at 70°C for 10 mins. Reaction mixture aliquots (cDNA) (2μl) were used as
templates for RT-PCR. PCR cycling conditions were 95°C for 15 min and 40 cycles of 95°C for
15 s, 60°C for 30 s, and 72°C for 30 s, followed by the final melting curve program. β-actin
RNA was used as the loading control. Each sample was done in triplicate, and the mean values
were used for quantization. The primers for HIF-1α, SCF, and β-actin were as follows: forward
5'-GCAAGCCCTGAAAGCG-3' and reverse 5'-GGCTGT CCGACTTTGA-3' (HIF-1α); for-
ward 5'-CTGCTCCTATTTAATCCTCTCGTCA-3' and reverse 5'-ATTGTACTACCATCTC
GCTTATCCA-3' (SCF); forward 5'-CAGAGCAAGAGAGGCATCC-3' and reverse 5'-CTGG
GGTGTTGA AGGTCTC (β-actin).
Chromatin immunoprecipitation assay (ChIP)
A commercial chip assay kit (Upstate Biotechnology, Waltham, USA) was used following the
manufacturers instructions. After treatment, each sample group was incubated with 1% form-
aldehyde to cross-link the DNA-protein complexes. The cross-links were heated at 37°C for
10 min. After being washed with cold PBS, the cells were collected and lysed with SDS lysis buffer
(1% SDS, 10 mM EDTA, 50 mM Tris, pH 8.1) containing protease inhibitors. Lysate was sonicat-
ed to shear the DNA to lengths between 200 and 1000 bp. The cross-linked protein was then im-
munoprecipitated using mouse anti-human HIF-1αmonoclonal antibody (1:100) or non-
specific IgG antibody (as the negative control of the antibody, Sigma, USA). The DNA was ex-
tracted via the phenol chloroform method and precipitated by ethanol for PCR amplification.
Primers flanking the hypoxia response element (HRE) of the VEGF promoter were used as the
positive control. Meanwhile, primers that do not include HRE were used as a negative control.
Additionally, the input and anti-RNA polymerase were used as the other positive controls of
the experiment [5]. The forward and reverse primers were as follows: 5'-GCCTGCTTCTCGC
CTACC-3' and 5'-GAGCTCCAGCATATTGCACG-3' (SCF), amplified the following sequence
of genomic DNA: GCCTGCTTCTCGCCTACCCCGGGCTCCGGAAGGGAAGGAGGCGTG
TCCGGAGCAGGCGGGCGGGAACTGTATAAAAGCGCCGGCGGCTCAGCAGCCGGG
CTTCGCTCGCCGCCTCGCGCCGAGACTAGAAGCGCTGCGGGAAGCAGGGACAGTG
GAGAGGGCGCTGCGCTCGGGCTACCCAATGCGTGGACTATCTGCCGCCGCTGTTC
GTGCAATATGCTGGAGCTC (228bp); 5'-GCCTCTGTCTGCCCAGCTGC-3' and 5'-GTGG
AGCTGAGAACGGGAAGC-3' (VEGF). The sequences of the negative primers were as follows
(not including HRE): forward: 5'-CGAGACCGGCGGGAGG-3', reverse: 5-CGGAGCC
CGGGGTAGG-3'. The PCR products were separated using 1% agarose.
Transient transfection and luciferase assay
The cells were plated at a density of 5×10
5
cells/well in 6-well plates. The pcDNA3.1-HIF-1α
plasmids were prepared as previously described. The full-length SCF luciferase promoter
plasmids containing the lengths of SCF 5-flanking sequences (spanning from +184 to-2185)
were constructed and named pGL3-SCF. Mutant SCF promoter was constructed and named
pGL3-SCF-M. The constructs were mutated from GCGTG to GTAGA that contained the SCF
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 4/14
promoters but without the HRE site. We then transfected the pcDNA3.1-HIF-1αplasmids
(1 μg) with either pGL3-SCF (1 μg) or pGL3-SCF-M (1 μg) into PANC-1 cells. Renilla lucifer-
ase construct (10 ng) (pRL-SV40, Promega, USA) was co-transfected as the internal reference.
PGL3-Basic (Promega, USA) and pGL3-VEGF were chosen as the negative and positive con-
trol, respectively. After transfection for 48 hrs, the cells were incubated in hypoxia for 12 hrs.
The luciferase activity was then determined using the Dual-Luciferase Reporter Assay System
(Promega, USA). All data was normalized to Renilla luciferase expression. VEGF luciferase re-
porter construction (pGL3-VEGF) was used as the positive control for the HIF-1αresponse.
Colony formation assay
The cells were seeded in six-well plates at a density of 200 cells per well and cultured at 37°C
for two weeks. At the end of the incubation period, colonies were stained with a three-step
stain set (Thermo Scientific) and counted using an optical microscope. Each measurement was
performed in triplicate, and each experiment was conducted at least three times.
Cell scratch assays
The pancreatic cells with different treatment were seeded to full confluency in 6-well plates
overnight. The following day, a scratch was introduced in the middle of the well by using a ster-
ile pipette tip. The medium was discarded and replaced with a fresh one. The rate of migration
towards the center of the wound was determined after 12 hrs.
Cell invasion assays
The invasion assays were performed with an 8.0 μm pore inserts in a 24-well Trans-well cham-
bers (Costar, USA). For this assay, 1×10
5
cells were isolated and added to the upper chamber of
a trans-well with DMEM. The invasion assay was performed using 1/6 diluted matrigel (BD
Bioscience)-coated filters. The DMEM with 10% fetal bovine serum was added to the lower
chamber and incubated for 24 hours. Cells that had migrated to the bottom of the filter were
stained with a three-step stain set (Thermo Scientific). Each experiment was repeated at least
three times.
Animal experiments
PANC-1 cells (1×10
6
) were injected subcutaneously into the right flank of female nude mice.
Digoxin and saline for injection were obtained from Tianjin Medical University Cancer Insti-
tute and Hospital. The cells were harvested by trypsinization, washed in PBS, resuspended at a
1:1 solution of PBS: Matrigel, and injected subcutaneously into the right flank of nude nu/nu
mice. When the tumors had grown to 200 mm
3
, the mice were randomized into two treatment
groups (n = 8 for each group): saline control and digoxin groups. The mice received daily intra-
peritoneal injections of either saline or digoxin (2 mg/kg) to inhibit HIF activity [2022]. The
tumor was palpable 5 days after inoculation, and all of the mice had developed tumors by the
end of the experiment. The physical condition of the animals, including fur-roughing, shed-
ding, and local trauma at the site of injection, as well as decrements in general animal activity
were regularly monitored. Tumor volume was calculated using the following formula: V = π/
6 × length × width
2
. The mice were immediately euthanized after the tumor samples were iso-
lated and photographed. All tumor samples were paraffin-embedded for IHC analysis.
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 5/14
Statistical analysis
All data analyses were performed using the SPSS13.0 statistical analysis software. Differences
between two samples were analyzed via unpaired t test. Multiple group comparisons were per-
formed via χ
2
test. Grade material related analysis was performed via Spearman's rank. Survival
analysis were performed via Kaplan-Meier and log-rank test. Differences were considered sta-
tistically significant at p <0.05.
Results
SCF and HIF-1αare over-expressed in PDAC and their expression
levels predict poor outcome
To investigate the relationship between HIF-1αand SCF in PDAC, we examined their expres-
sion pattern in human pancreatic adenocarcinoma tissue. IHC was performed on serial sec-
tions of 95 PDAC samples. We observed that HIF-1αand SCF have higher expression in
cancer samples (Fig. 1A). The Spearman analysis showed that an obvious correlation exists be-
tween SCF and HIF-1α(r = 0.728, p<0.001: S1 Table). The SCF level was correlated with the
lymph node metastasis (r = 0.281, p<0.05: Table 1) and the pathological tumor node metastasis
(pTNM) stages (r = 0.353, p<0.05) in these cancer samples. The results showed that HIF-1αor
SCF higher-expression group had significantly lower survival rate than the HIF-1αor SCF
lower-expression group (p<0.05, respectively: Fig. 1B and C).
These results indicate that SCF and HIF-1αwere over-expressed in PDAC, and their expres-
sion levels can predict poor outcome.
Fig 1. HIF-1αand SCF expression in pancreatic adenocarcinoma tissue and the cumulative survival analysis. (A): The IHC results of HIF-lαand SCF.
SCF protein expression was significantly correlated with HIF-lα, as detected by immunohistochemical staining on PDAC. Left panels: various expression
levels of HIF-lαprotein. Right panels: expression of SCF protein of the same samples in the adjacent section. (B): The results of survivalanalysis on HIF-lα.
PDAC patients (n = 95) with high positive HIF-lαprotein expression had significantly worse total survival than those with low or medium positive expression.
(C): The results of survival analysis on SCF. The same PDAC patients (n = 95) with high positive SCF protein expression had significantly worse total survival
than those with low or medium positive expression.
doi:10.1371/journal.pone.0121338.g001
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 6/14
Expression of HIF-1αand SCF in pancreatic cancer cells under hypoxia
The relationship between SCF and HIF-1αwas further investigated under hypoxia in vitro.
The results revealed that two pancreatic cancer cell lines, PANC-1 and BxPC-3, expressed little
HIF-1αand SCF protein under normoxia, and their expressions were obviously increased after
hypoxia treatment, especially at 12 hrs (Fig. 2A and B). As a positive control, VEGF expression
increased correspondingly (data not show). This result indicates that hypoxia up-regulated the
HIF-1αand SCF protein expression in PANC-1 and BxPC-3 pancreatic cancer cell lines. To in-
vestigate whether HIF-1αregulate the expression of SCF, siRNA targeting HIF-1αwas used.
The two pancreatic cancer cell lines were transfected with siRNA targeting HIF-1αand cul-
tured under hypoxia (1% O
2
) for 12 hrs (defined as Hsi, the same below), and the two cell lines
only cultured under hypoxia (1% O
2
) for 12 hrs were used as the control sample (defined as
HC, the same below), as shown in Fig. 2C and D. After interference in PANC-1 and BxPC-3
cell lines, HIF-1αand SCF protein levels decreased evidently compared with those of control
groups. The results indicate that siRNA targeting of HIF-1αpartially or completely block the
expression of SCF under hypoxia. The SCF and HIF-1αmRNA expressions were then tested
via quantitative RT-PCR after hypoxia treatment or siRNA targeting HIF-1αtransfection cul-
ture. The results indicate that both mRNA levels of HIF-1αand SCF increased or decreased
with the same trend (Fig. 2E, F, G and H): In PANC-1 cells, compared with normoxia control,
HIF-1αexpression was increased for 8.1 fold and SCF was increased for 10.2 fold because of
hypoxia treatment, and in BxPC-3 cells, HIF-1αexpression was increased for 10.5 fold and
SCF was increased for 7.3 fold because of hypoxia treatment. After HIF-1αknockdown at hyp-
oxia environment in PANC-1 cells, HIF-1αexpression was reduced to 84% and SCF was re-
duced to 80%. In BxPC-3 cells, HIF-1αexpression was reduced to 78% and SCF was reduced to
81%. These results indicate that the up-regulation or suppression of SCF expression is directly
related on the regulation of HIF-lα.
Table 1. Correlations between clinicopathological features and SCF expression in PDAC.
SCF
Low Medium High r
s
p-values
Age
a
59 (3679) 5.747
a
0.06
Tumor size (cm)
b
3.649±1.67 4.435±1.359 6.107±2.99 10.809
b
<0.001
Histological grade 0.073 0.48
G1 16 13 10
G2 9 6 6
G3 14 7 14
LN metastasis 0.281 0.006
N 0 22 11 7
N 1 17 15 23
pTNM stage 0.353 0
I,II 23 13 6
III 9 5 8
IV 7 8 16
*p-values were calculated by Spearman's Rank-Correlation test (n = 95)
Age
a
: Expressed as median (range), χ
2
= 5.747, p = 0.060 (kruskal-wallis test)
Tumor size (cm)
b
: Expressed as mean, F = 10.809, p<0.001 (anova test)
doi:10.1371/journal.pone.0121338.t001
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 7/14
HIF-1αdirectly binds to the SCF promoter and up-regulated the SCF
promoter activity
After the screening the 5-flanking region of the SCF gene, we found a potential HIF-1 binding
site(-68 *-64)near the start site of transcription (Fig. 3A). To further investigate the relation-
ship between SCF and HIF-1αin PDAC cell lines and to validate the binding of HIF-lαto the
SCF promoter, an independent ChIP assay was performed after 12 hrs of hypoxia treatment.
The promoter fragment was specially co-immunoprecipitated by HIF-lαantibody but not the
negative controls (Fig. 3B). Furthermore, the SCF promoter region (-1163 to-955), which did
not contain HRE, cannot be pulled down by the HIF-1αantibody, thereby suggesting that
HIF-1αdirectly binds to the HRE region of the SCF promoter in vitro. To determine whether
Fig 2. Expression of HIF-1αand SCF protein and mRNA after different treatment. (A, B): Expression of HIF-1αand SCF protein after hypoxia treatment.
Two pancreatic cancer cell lines, PANC-1 and BxPC-3, were incubated under hypoxia at different time points (0, 3, 6, 12 hrs). Both of the protein expression
of HIF-1αand SCF were determined via Western blot. β-actin expression was used as the control. (C, D): Expression of HIF-1αand SCF protein after
transfected with siRNA targeting HIF-1α. The PANC-1 and BxPC-3 cancer cells were transfected with siRNA targeting HIF-1αfor 48 hrs and incubated under
hypoxia for 12 hrs. The protein expressions of HIF-1αand SCF were then determined via Western blot. (E, F, G, H): Expression of HIF-1αand SCF mRNA
after different treatments. After hypoxia treatment or interference of siRNA-HIF-1αtreatment in PANC-1 and BxPC-3 cancer cells, the mRNA expressions
were quantified via real-time PCR. Data from three experimental determinations and bars indicate the SD, p <0.05 vs. control.
doi:10.1371/journal.pone.0121338.g002
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 8/14
the binding of HIF-1αto the promoter can activate SCF, transient transfection and dual lucif-
erase assay was performed to detect the SCF promoter activity after HIF-1αover-expression.
The results show that the full-length SCF promoter activity (pGL3-SCF) was increased
12.8-fold after the over expression of HIF-1αcompared to that treated with plasmids alone,
but the mutation of SCF promoters (pGL3-SCFM) did not increase (p<0.05: Fig. 3C). Over-ex-
pression of HIF-1αwas increased 11.6-fold of the VEGF promoter activity compared to the
controls (p<0.05: Fig. 3C). Thus, HIF-1αdirectly transactivates SCF expression.
Influence of SCF on the biological behaviors of PDAC cell lines
To investigate the influence of SCF on the biological behaviors of PDAC cell lines, exogenous
SCF (150 ng/ml, the same below) or SCF neutralizing antibody (10 ng/ml, the same below) and
siRNA was used to increase or decrease the expression of SCF in PANC-1 cell lines. Colony for-
mation was used to detect cell proliferation ability and cell scratch or/and transwell chambers
experiments were used to detect cell invasion.
After applying the interference of exogenous SCF and SCF neutralizing antibody or siRNA
into PANC-1 cells, the proliferative ability in the interference groups significantly increased
and decreased compared with those of the control groups, respectively (p<0.05, respectively:
Fig. 4A and B). Similarly, after the interference of exogenous SCF and anti-SCF neutralizing
antibody, the scratch space at 12 hrs of PANC-1 cells was detected. The widths in the SCF or
anti-SCF groups were smaller or bigger than those in the NC or HC control group (p <0.05,
respectively: Fig. 4C). Moreover, after the interference of exogenous SCF and anti-SCF
Fig 3. HIF-1αdirectly binds to the HRE region of SCF promoter and up-regulates the activity. (A): The DNA sequence of the SCF promoter. A potential
HIF-1αbinding site is located at 68 to 64 near the start site of transcription. (B): Chromatin immunoprecipitation analysis. PANC-1 cells were cultured
under normoxia and hypoxia for 12 h and then analyzed. The PCR products of SCF promoter were only detected in the samples that were precipitated by
HIF-1αantibodies but not in control IgG samples. The SCF promoter region (-1163 to-955) that do not contain HRE (non-HRE) cannot be precipitated by HIF-
1αantibodies. (C): Dual luciferase results in different groups. After transfected with pcDNA3.1-HIF-1αplasmids (1 μg) with pGL3-SCF (1μg) or
(pGL3-SCF-M) (1μg), pGL3-VEGF was used as the positive control. After transfection incubation for 48 h and incubated under hypoxia for 12 hrs, the cells
were harvested for dual luciferase assay. The results showed that the full-length SCF promoter activity (pGL3-SCF) was increased for 12.8 fold after over
expression of HIF-1αcompared to that treated with plasmids alone, but the mutation of SCF promoters (pGL3-SCF-M) did not increase. Over-expression of
HIF-1αwas increased for 11.6 fold of the VEGF promoter activity compared to the controls.
doi:10.1371/journal.pone.0121338.g003
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 9/14
neutralizing antibody, the number of cells coming through the membrane was significantly
more and less than that of the control group, respectively (p<0.05, respectively: Fig. 4D).
HIF-1αpromote the development of PDAC by regulating SCF
The results described above pointed out a possibility that HIF-1a may promote PDAC progres-
sion by regulating SCF. We rescued the cell proliferation and invasion abilities by adding exog-
enous SCF when transfected with siRNA targeting HIF-1αat the same time (Fig. 5A). Colony
formation results showed that the decreased tumor cells growth rate following HIF-1αknock-
down can be rescued by the up-regulation of SCF (p<0.05, Fig. 5B). Similarly, both cell scratch
and transwell chambers results showed that the decreased tumor cell invasion rate following
HIF-1αknockdown can be rescued by adding exogenous SCF (p<0.05, respectively: Fig. 5C
and D).
Fig 4. Influence of SCF on the biological behavior of PDAC cell. (A) and (B): Colony formation results showed the SCF effect on cell proliferation. After
applying the interference of exogenous SCF or anti-SCF antibody in PANC-1 cells, the proliferative ability in the interference groups significantly increased or
decreased compared with those of the control groups (p<0.05, respectively). *stands for p<0.05 (mark group VS control group). (C): The scratch results
showed the SCF effect on cell invasion. After the interference of exogenous SCF and anti-SCFantibody, the widths in the SCF or anti-SCF groups were
smaller or bigger than those in the NC or HC control group (p <0.05, respectively). (D): The transwell chambers results showed the SCF effect on cell
invasion. After the interference of exogenous SCF or anti-SCF, the cell number through the membrane was significantly more or less than the control group
(p <0.05, respectively). *stands for p<0.05 (N+SCF group VS NC group), and # stands for p<0.05 (H +anti-SCF group VS HC group).
doi:10.1371/journal.pone.0121338.g004
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 10 / 14
Taken together, these results indicate that SCF, a downstream gene of HIF-1α, can promote
the development of PDAC in vitro.
Inhibiting effect of digoxin against PDAC in vivo
To determine whether HIF-1αregulate SCF expression in vivo, we injected PANC-1 cells sub-
cutaneously into the right flank of nude nu/nu mice. The results were as follows: compared
with the saline control group, the average tumor volume in digoxin group was reduced signifi-
cantly (p<0.05, Fig. 6A). Next, we evaluated the correlation between HIF-1αand SCF in the
mice tumors by IHC, and the results suggest that expression of SCF was decreased as a result of
the HIF-1αlevel reduction by digoxin (Fig. 6B). Treatment with digoxin resulted in no signifi-
cant difference in the body weight of treated mice, none of the tested mice manifested signs of
other adverse effects as specified in the method section, and no toxicity on the blood count or
hepatic and renal function was observed with digoxin treatment (data not shown). These re-
sults indicate the anti-tumor effect and safety of digoxin in vivo.
Fig 5. PDAC cells proliferation and invasion abilities can be rescued by up-regulation SCF followingHIF-1αknockdown. (A): The levels of HIF-1α
and SCF protein were tested via Western blot in different groups: The SCF protein level can be rescued by adding exogenous SCF when transfected with
siRNA targeting HIF-1αat the same time. (B): Colony formation results in different groups: the decreased tumor cells proliferation rate following HIF-1α
knockdown can be rescued by up-regulation of SCF (p<0.05, respectively). (C): The scratch results in different groups: the decreased tumor cells
proliferation rate following HIF-1αknockdown can be rescued by up-regulation of SCF (p<0.05, respectively). (D): Transwell chambers results in different
groups: the decreased tumor cells through the membrane following HIF-1αknockdown can be rescued by up-regulation of SCF (p<0.05, respectively). *
stands for p<0.05 (siRNA-HIF-1αgroup VS control group), and
#
stands for p<0.05 (siRNA-HIF-1α+SCF group VS siRNA-HIF-1αgroup).
doi:10.1371/journal.pone.0121338.g005
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 11 / 14
Discussion
In this report, we proved that HIF-1αand SCF were prognostic factors in PDAC by examining
the expression of SCF and HIF-1αin pancreatic cancer tissues and analyzing the clinical fea-
tures and prognosis. A strong correlation was observed between the expressions of these two
factors in primary PDAC tissues. We also found that HIF-1αpromoted the development of
PDAC by trans-activating SCF under hypoxia in vitro. Furthermore, SCF promoted PDAC cell
proliferation and invasion, and the decreased tumor cell proliferation and invasion abilities fol-
lowing HIF-1αknockdown can be rescued by the up-regulation of SCF under hypoxia. SCF
has been suggested as a direct target gene of HIF-1 in MCF-7 cells through in vitro analysis [5].
We extended this effect in pancreatic cancers with more consolidated evidence including clini-
cal data as well as in vitro and in vivo analysis, thereby suggesting that the regulatory role of
HIF-1 on SCF is general in cancer cells. Interestingly, we recently identified SCF as an indepen-
dent predictor for poor prognosis of patients with hepatocellular carcinoma [23]. In accor-
dance with this study, SCF expression was associated with a shorter survival rate in resected
patients. For those patients with high SCF expression, strengthened adjuvant therapy should
be given to achieve a better outcome. Another study reported that serum analysis of PDAC
also showed elevated levels of SCF in pretreated patients with pancreatic cancer [24], thereby
suggesting that SCF could also be used as a diagnostic marker for pancreatic cancer. Accumu-
lating evidences have confirmed that HIF-1 had crucial functions in the pathogenesis of pan-
creatic cancer. The present study further demonstrated that the proliferative and invasive effect
of HIF-1 was, at least in part, mediated by SCF. Previous studies have shown that SCF activated
the expression of HIF-1 in pancreatic cancer [11]. We postulate that a positive feedback loop
exists between SCF and HIF. The loop may function to maintain the constitutive expression of
HIF-1 under normoxia and strengthen the pathogenic effect of SCF under hypoxia.
SCF exerted its biological functions through binding to a specific ligand, c-kit. C-kit appears
in embryonic tissues of human beings but was silenced in adult tissues. Recent clinical evidence
showed that c-kit was negative in normal pancreatic tissues but is over-expressed in malignant
pancreatic tissues, including cancerous duct, exocrine pancreas and βcells of islet, thereby sug-
gesting that the SCF/c-kit pathway might be involved in the malignant transformation of pan-
creatic cells [25]. As shown in the present study, SCF mainly promoted invasion and
proliferation of pancreatic cancer cells. The hypoxia-induced SCF expression might further
Fig 6. Inhibiting effect of digoxin against PDAC in vivo. (A): The mice tumorsvolume in different groups. After six weeks of treatment, digoxin therapy
decreased the tumorsvolume, which was significantly smaller than those in the saline control group (p<0.05). (B): IHC results in different groups.
Additionally, the IHC results confirmed the effect of digoxin: Digoxin therapy decreased the HIF-lαand SCF positive tumor cells compared to the saline
control group, respectively.
doi:10.1371/journal.pone.0121338.g006
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 12 / 14
accelerate the progression of pancreatic cancer because hypoxia is a typical microenvironment
of pancreatic cancer. Another interesting function of SCF is to regulate the differentiation of
PANC-1 pancreatic cancer cells into insulin-producing cells. We postulate that hypoxia might
contribute to the maintenance of endocrine function of malignant pancreatic tissues through
SCF/c-kit pathway.
Based on the findings of this study, we suggest that the SCF/c-kit pathway may be a poten-
tial target for the treatment of pancreatic cancer. Thus far, several c-kit inhibitors such as Ima-
tinib and Sunitinib have been approved by FDA to the treatment of leukemia, renal cancer cell,
and gastrointestinal stromal tumors [26]. Further clinical trial should be performed to use
these reagents, alone or in combination with gemcitabine, as new strategies for the treatment of
pancreatic cancer.
In conclusion, we identified SCF as a direct target of HIF-1 in pancreatic cancer. SCF/c-kit
promoted invasion and proliferation of pancreatic cancer cells and may be a new target for the
treatment of pancreatic cancer.
Supporting Information
S1 Table. Correlations between HIF-1αand SCF expression in pancreatic cancer. Statistical
analysis of immunohistochemical results of HIF-1αand SCF expression in human PDAC sur-
gical samples. p values were analyzed by Spearmans rank-correlation test.
(DOC)
Author Contributions
Conceived and designed the experiments: CG SL TZ JC HR JH. Performed the experiments:
CG SL TZ JC HR HZ XW ML. Analyzed the data: CG SL TZ JC HR HZ XW ML JL SG XZ JS
ZY JH. Contributed reagents/materials/analysis tools: CG SL TZ JC HR HZ XW ML JL SG XZ
JS ZY JH. Wrote the paper: CG SL TZ JC HR ML JH.
References
1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013; 63:1130. doi: 10.
3322/caac.21166 PMID: 23335087
2. Gu Y, Runyan C, Shoemaker A, Surani MA, Wylie C. Membrane-bound steel factor maintains a high
local concentration for mouse primordial germ cell motility, and defines the region of their migration.
PloS one. 2011; 6:e25984. doi: 10.1371/journal.pone.0025984 PMID: 21998739
3. Bai CG, Hou XW, Wang F, Qiu C, Zhu Y, Huang L, et al. Stem cell factor-mediated wild-type KIT recep-
tor activation is critical for gastrointestinal stromal tumor cell growth. World journal of gastroenterology:
WJG. 2012; 18:29292937. doi: 10.3748/wjg.v18.i23.2929 PMID: 22736916
4. Talaiezadeh A, Jazayeri SN, Nateghi J. Expression of c-kit protein in cancer vs. normal breast tissue.
Contemporary oncology. 2012; 16:306309. doi: 10.5114/wo.2012.30058 PMID: 23788899
5. Han ZB, Ren H, Zhao H, Chi Y, Chen K, Zhou B, et al. Hypoxia-inducible factor (HIF)-1 alpha directly
enhances the transcriptional activity of stem cell factor (SCF) in response to hypoxia and epidermal
growth factor (EGF). Carcinogenesis. 2008; 29:18531861. doi: 10.1093/carcin/bgn066 PMID:
18339685
6. Pedersen M, Lofstedt T, Sun J, Holmquist-Mengelbier L, Pahlman S, Ronnstrand L. Stem cell factor in-
duces HIF-1alpha at normoxia in hematopoietic cells. Biochemical and biophysical research communi-
cations. 2008; 377:98103. doi: 10.1016/j.bbrc.2008.09.102 PMID: 18834862
7. Gibbs BF, Yasinska IM, Oniku AE, Sumbayev VV. Effects of stem cell factor on hypoxia-inducible factor
1 alpha accumulation in human acute myeloid leukaemia and LAD2 mast cells. PloS one. 2011; 6:
e22502. doi: 10.1371/journal.pone.0022502 PMID: 21799876
8. Arko L, Katsyv I, Park GE, Luan WP, Park JK. Experimental approaches for the treatment of malignant
gliomas. Pharmacology & therapeutics. 2010; 128:136.
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 13 / 14
9. Heissig B, Werb Z, Rafii S, Hattori K. Role of c-kit/Kit ligand signaling in regulating vasculogenesis.
Thrombosis and haemostasis. 2003; 90:570576. PMID: 14515175
10. Lennartsson J, Jelacic T, Linnekin D, Shivakrupa R. Normal and oncogenic forms of the receptor tyro-
sine kinase kit. Stem cells. 2005; 23:1643. PMID: 15625120
11. Zhang M, Ma Q, Hu H, Zhang D, Li J, Ma G, et al. Stem cell factor/c-kit signaling enhancesinvasion of
pancreatic cancer cells via HIF-1alpha under normoxic condition. Cancer letters. 2011; 303:108117.
doi: 10.1016/j.canlet.2011.01.017 PMID: 21320746
12. Nogueira V, Park Y, Chen CC, Xu PZ, Chen ML, Tonic I, et al. Akt determines replicative senescence
and oxidative or oncogenic premature senescence and sensitizes cells to oxidative apoptosis. Cancer
cell. 2008; 14:458470. doi: 10.1016/j.ccr.2008.11.003 PMID: 19061837
13. Krock BL, Skuli N, Simon MC. Hypoxia-induced angiogenesis: good and evil. Genes & cancer. 2011;
2:11171133.
14. Miyake K, Yoshizumi T, Imura S, Sugimoto K, Batmunkh E, Kanemura H, et al. Expression of hypoxia-
inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreaticcarci-
noma: correlation with poor prognosis with possible regulation. Pancreas. 2008; 36:e19. doi: 10.1097/
MPA.0b013e3181675010 PMID: 18437073
15. Zhu Z, Wang Y, Liu Z, Wang F, Zhao Q. Inhibitory effects of epigallocatechin-3-gallate on cell prolifera-
tion and the expression of HIF-1alpha and P-gp in the human pancreatic carcinoma cell line PANC-1.
Oncology reports. 2012; 27:15671572. doi: 10.3892/or.2012.1697 PMID: 22367292
16. Miyake K, Nishioka M, Imura S, Batmunkh E, Uto Y, Nagasawa H, et al. The novel hypoxic cytotoxin,
TX-2098 has antitumor effect in pancreatic cancer; possible mechanism through inhibiting VEGF and
hypoxia inducible factor-1alpha targeted gene expression. Experimental cell research. 2012;
318:15541563. doi: 10.1016/j.yexcr.2012.03.013 PMID: 22472348
17. Knaup KX, Jozefowski K, Schmidt R, Bernhardt WM, Weidemann A, Juergensen JS, et al. Mutual regu-
lation of hypoxia-inducible factor and mammalian target of rapamycin as a function of oxygen availabili-
ty. Molecular cancer research: MCR. 2009; 7:8898. doi: 10.1158/1541-7786.MCR-08-0288 PMID:
19147540
18. Seenath MM, Roberts D, Cawthorne C, Saunders MP, Armstrong GR, O'Dwyer ST, et al. Reciprocal re-
lationship between expression of hypoxia inducible factor 1alpha (HIF-1alpha) and the pro-apoptotic
protein bid in ex vivo colorectal cancer. British journal of cancer. 2008; 99:459463. doi: 10.1038/sj.bjc.
6604474 PMID: 18648372
19. Hoffmann AC, Mori R, Vallbohmer D, Brabender J, Klein E, Drebber U, et al. High expression of HIF1a
is a predictor of clinical outcome in patients with pancreatic ductal adenocarcinomas and correlated to
PDGFA, VEGF, and bFGF. Neoplasia. 2008; 10:674679. PMID: 18592007
20. Chaturvedi P, Gilkes DM, Wong CC, Kshitiz, Luo W, Zhang H, et al. Hypoxia-inducible factor-depen-
dent breast cancer-mesenchymal stem cell bidirectional signaling promotes metastasis. The Journal of
clinical investigation. 2013; 123:189205. doi: 10.1172/JCI64993 PMID: 23318994
21. Fujisawa T, Joshi BH, Puri RK. Histone modification enhances the effectiveness of IL-13 receptor tar-
geted immunotoxin in murine models of human pancreatic cancer. Journal of translational medicine.
2011; 9:37. doi: 10.1186/1479-5876-9-37 PMID: 21477288
22. Zhang H, Wong CC, Wei H, Gilkes DM, Korangath P, Chaturvedi P, et al. HIF-1-dependent expression
of angiopoietin-like 4 and L1CAM mediates vascular metastasis of hypoxic breast cancer cells to the
lungs. Oncogene. 2012; 31:17571770. doi: 10.1038/onc.2011.365 PMID: 21860410
23. Wang X, Ren H, Zhao T, Chen J, Sun W, Sun Y, et al. Stem cell factor is a novel independent prognos-
tic biomarker for hepatocellular carcinoma after curative resection. Carcinogenesis. 2014; 35:2283
2290. doi: 10.1093/carcin/bgu162 PMID: 25086759
24. Torres C, Perales S, Alejandre MJ, Iglesias J, Palomino RJ, Martin M, et al. Serum cytokine profile in
patients with pancreatic cancer. Pancreas. 2014; 43:10421049. doi: 10.1097/MPA.
0000000000000155 PMID: 24979617
25. Amsterdam A, Raanan C, Polin N, Melzer E, Givol D, Schreiber L. Modulation of c-kit expression in pan-
creatic adenocarcinoma: a novel stem cell marker responsible for the progression of the disease.Acta
histochemica. 2014; 116:197203. doi: 10.1016/j.acthis.2013.07.002 PMID: 23978330
26. Schallier D, Trullemans F, Fontaine C, Decoster L, De Greve J. Tyrosine kinase inhibitor-induced mac-
rocytosis. Anticancer research. 2009; 29:52255228. PMID: 20044640
SCF Promotes Pancreatic Ductal Adenocarcinoma Cell Progression
PLOS ONE | DOI:10.1371/journal.pone.0121338 March 23, 2015 14 / 14
... The binding of stem cell factor (SCF) to c-Kit (also known as CD117) leads to the activation of multiple signaling pathways, including the phosphoinositide 3 kinase, phospholipase C-gamma, Src kinase, Janus kinase-signal transducer and activator of transcription, c-Kit kinase activity is dysregulated in various pathological conditions, including cancer, allergy, and vascular leakage [4][5][6][7][8]. Previous studies have reported that the expression of c-Kit with oncogenic mutations is dysregulated or upregulated in various cancers, which results in SCF-independent c-Kit activation and cell proliferation. ...
... Previous studies have reported that the expression of c-Kit with oncogenic mutations is dysregulated or upregulated in various cancers, which results in SCF-independent c-Kit activation and cell proliferation. Overexpression of c-Kit is induced by hypoxia as well as activation of various transcription factors, including AP-2, ETS, SP1, MYB, and MITF [7,[9][10][11][12][13][14][15]. Currently, more than 500 c-Kit mutations have been identified in human tumors (Sanger Institute Catalogue of Somatic Mutations in Cancer, https://cancer.sanger.ac.uk). ...
Article
Full-text available
c‐Kit overexpression and activating mutations, which are reported in various cancers, including gastrointestinal stromal tumor (GIST), small cell lung cancer (SCLC), acute myeloid leukemia, acral melanoma, and systemic mastocytosis (SM), confer resistance to tyrosine kinase inhibitors (TKIs). To overcome TKI resistance, an anti‐c‐Kit antibody–drug conjugate was developed in this study to treat wild‐type and mutant c‐Kit‐positive cancers. NN2101, a fully human IgG1, was conjugated to DM1, a microtubule inhibitor, through N‐succinimidyl‐4‐(N‐maleimidomethyl) cyclohexane‐1‐carboxylate (SMCC) (to give NN2101‐DM1). The antitumor activity of NN2101‐DM1 was evaluated in vitro and in vivo using various cancer cell lines. NN2101‐DM1 exhibited potent growth‐inhibitory activities against c‐Kit‐positive cancer cell lines. In a mouse xenograft model, NN2101‐DM1 exhibited potent growth‐inhibitory activities against imatinib‐resistant GIST and SM cells. In addition, NN2101‐DM1 exhibited a significantly higher anti‐cancer effect than carboplatin/etoposide against SCLC cells where c‐Kit does not mediate cancer pathogenesis. Furthermore, the combination of NN2101‐DM1 with imatinib in imatinib‐sensitive GIST cells induced complete remission compared to treatment with NN2101‐DM1 or imatinib alone in mouse xenograft models. These results suggest that NN2101‐DM1 is a potential therapeutic agent for wild‐type and mutant c‐Kit‐positive cancers.
... A HIF-1 binding site has been identified and characterized in the VEGF gene, and ARNT-deleted cells failed to increase VEGF expression levels upon exposure to hypoxic stimuli 114 . Additionally, other angiogenic growth factors, including stromal-derived factor 1, stem cell factor, and angiopoietin family members, are also regulated by HIFs and participate in tumor vascularization [115][116][117] . ...
Article
Full-text available
Oxygen is crucial for life and acts as the final electron acceptor in mitochondrial energy production. Cells adapt to varying oxygen levels through intricate response systems. Hypoxia-inducible factors (HIFs), including HIF-1α and HIF-2α, orchestrate the cellular hypoxic response, activating genes to increase the oxygen supply and reduce expenditure. Under conditions of excess oxygen and resulting oxidative stress, nuclear factor erythroid 2-related factor 2 (NRF2) activates hundreds of genes for oxidant removal and adaptive cell survival. Hypoxia and oxidative stress are core hallmarks of solid tumors and activated HIFs and NRF2 play pivotal roles in tumor growth and progression. The complex interplay between hypoxia and oxidative stress within the tumor microenvironment adds another layer of intricacy to the HIF and NRF2 signaling systems. This review aimed to elucidate the dynamic changes and functions of the HIF and NRF2 signaling pathways in response to conditions of hypoxia and oxidative stress, emphasizing their implications within the tumor milieu. Additionally, this review explored the elaborate interplay between HIFs and NRF2, providing insights into the significance of these interactions for the development of novel cancer treatment strategies.
... We did not detect IL-6, IL-1 α, or IL-8 in our study, which could be due to the sample size. In addition, previous studies have found SCF to be elevated in pancreatic ductal adenocarcinoma patients, and SCF plays an important role in the pathophysiology of mast cells [67,68]. Other cytokines (IL-1β, BCA-1, TNF-α, CRP, ENA-78, MDC, and TNFRII) showed no obvious differences between the non-smoker (n = 30), current smoker (n = 30), and former smoker groups (n = 18). ...
Article
Full-text available
Cardiovascular and respiratory diseases, and several cancers resulting from tobacco smoking, are initially characterized by chronic systemic inflammation. Cytokine imbalances can result in inflammation, making it important to understand the pathological mechanisms behind cytokine production. In this study, we collected blood samples from 78 healthy male volunteers, including non-smokers (n = 30), current smokers (n = 30), and ex-smokers (n = 18), and utilized the liquid suspension chip technique to investigate and compare the expression levels of 17 cytokines and chemokines in the human serum of these volunteers. The results demonstrated that the expression levels of CXCL9/MIG and sIL-6R significantly increased after smoking, and continued to increase after quitting smoking. The expression levels of TARC, ITAC, and sVEGFR-3 increased after smoking but decreased after quitting smoking; the expression level of SAA significantly decreased after smoking and showed an upward trend after quitting smoking. Seven cytokines (IL-1β, BCA-1, TNF-α, CRP, ENA-78, MDC, and TNFRII) did not vary between the three groups, while four cytokines (IL-1α, IL-6, IL-8, and SCF) were not detected in any serum sample. In conclusion, this study assessed the physiological production of cytokines and chemokines, highlighting the differences in each due to smoking status. Our results could help evaluate the early development of smoking-related chronic diseases and cancers.
... En effet, une augmentation de densité mastocytaire a été observée chez le rat en période néonatale dans l'hémisphère cérébral exposé à une hypoxie (Jin et al., 2007). Ce résultat peut être expliqué par la présence dans le promoteur du gène SCF d'éléments de réponse au facteur HIF-1α, élément régulateur central de l'hypoxie (Gao et al., 2015). De plus, l'hypoxie peut entrainer une augmentation d'expression du récepteur du SCF, KIT, notamment dans des cellules endothéliales (Kim et al., 2019). ...
Thesis
La mise en place de la voie de synthèse des minéralocorticoïdes au cours du développement foetal chez l’homme reste un processus encore non totalement élucidé, que ce soit du point de vue de la chronologie d’expression des enzymes de la stéroïdogenèse ou bien des acteurs déterminant la production d’aldostérone en période péri-natale. Chez l’adulte, la sécrétion d’aldostérone est contrôlée majoritairement par des facteurs circulants tels que le système rénine angiotensine et le potassium. L’importance des mécanismes paracrines a été plus récemment démontré, impliquant différents acteurs, dont les mastocytes intra-surrénaliens situés dans la région sous-capsulaire. En effet, les produits de sécrétion mastocytaire stimulent la production d’aldostérone par l’intermédiaire de la libération de sérotonine et de l’activation des récepteurs sérotoninergiques de type 4 présents à la surface des cellules stéroïdogènes. Cependant, le rôle des mastocytes et de la voie sérotoninergique dans le développement surrénalien n’a jamais été étudié. Dans ce travail, nous montrons que les mastocytes colonisent la surrénale foetale vers 18 semaines d’aménorrhée (SA) dans la zone définitive de façon conjointe à l’apparition des premières cellules exprimant la 3β-hydroxystéroïde déshydrogénase. L’aldostérone synthase (CYP11B2) est détectée dans la même zone vers 24 SA, puis son expression augmente au cours du développement. De façon intéressante, nous mettons en évidence une densité mastocytaire ainsi qu’une expression de CYP11B2 plus importantes chez les foetus comparés aux nouveau-nés de termes équivalents, suggérant l’implication des mastocytes dans le processus d’adaptation à la vie extra-utérine. Par ailleurs, nous rapportons une activation des voies de signalisation Wnt/β-caténine et Shh au cours du développement surrénalien, qui ne semble pas contrôler la prolifération cellulaire. La voie Shh pourrait être associée à l’acquisition du phénotype stéroïdogène mature alors que la voie β-caténine qui est active antérieurement à l’expression de CYP11B2 ne semble pas suffisante pour initier la fonction minéralocorticoïde. Globalement, ce travail montre la chronologie de la différenciation de la zone définitive surrénalienne au cours du développement et révèle pour la première fois la mise en place de système de régulation paracrine impliquée dans la production d’aldostérone en anté et post-natal.
... 18 HIF-1α is a helix-loop-helix transcription factor that drives the expression of many genes vital in all aspects of the cancer cell invasion and metastasis. [19][20][21][22][23][24][25][26][27][28][29][30][31] Multiple studies of HIF-1α and PDAC have shown a significant association between HIF-1α overexpression and poor prognosis. [32][33][34][35] High HIF-1α expression in PDAC is a predictor for metastatic disease. ...
Article
Full-text available
The Cancer Genome Atlas (TCGA) of a pancreatic cancer cohort identified high MST1R (RON tyrosine kinase receptor) expression correlated with poor prognosis in human pancreatic cancer. RON expression is null/minimal in normal pancreas but elevates from pan‐in lesions through invasive carcinomas. We report using multiple approaches RON directly regulates HIF‐1α, a critical driver of genes involved in cancer cell invasion and metastasis. RON and HIF‐1α are highly co‐expressed in the 101 human PDAC tumors analyzed and RON expression correlated with HIF‐1α expression in a subset of PDAC cell lines. knockdown of RON expression in RON positive cells blocked HIF‐1α expression, whereas ectopic RON expression in RON null cells induced HIF‐1α expression suggesting the direct regulation of HIF‐1α by RON kinase receptor. RON regulates HIF‐1α through an unreported transcriptional mechanism involving PI3 kinase‐mediated AKT phosphorylation and Sp1‐dependent HIF‐1α promoter activity leading to increased HIF‐1α mRNA expression. RON/HIF‐1α modulation altered the invasive behavior of PDAC cells. A small‐molecule RON kinase inhibitor decreased RON ligand, MSP‐induced HIF‐1α expression, and invasion of PDAC cells. Immunohistochemical analysis on RON knockdown orthotopic PDAC tumor xenograft confirmed that RON inhibition significantly blocked HIF‐1α expression. RON/HIF‐1α co‐expression also exists in triple‐negative breast cancer cells, a tumor type that also lacks molecular therapeutic targets. This is the first report describing RON/HIF‐1α axis in any tumor type and is a potential novel therapeutic target.
... [17][18][19] Also, some studies have reported the relation of SCF/c-Kit expression and tumor aggressiveness and patients' poor prognosis. 16,[20][21][22] In addition, activated SCF/c-Kit pathway is related to tumor resistance to treatment. [23][24][25] Sun et al. demonstrated that SCF expression by glioma cells can activate brain microvascular endothelial cells and consequently increase brain tumors angiogenesis in animal models. ...
Article
Full-text available
Background Investigation of novel blood-circulating agents as potential biomarkers for glioblastoma multiforme (GBM) patients’ diagnosis and monitoring has gained lots of attention, due to limitations of imaging modalities and invasive tissue biopsy procedures. The present study aims to assess the diagnostic and prognostic values of preoperative stem cell factor (SCF) plasma level in GBM patients. Methods Preoperative plasma samples from 58 GBM patients and 20 patients with nonglial tumors and 30 healthy controls were obtained. SCF levels were measured by employing the enzyme-linked immunosorbent assay test and the values were compared between these three groups. Then, the association of SCF plasma level and tumor volume, progression-free survival (PFS), and overall survival (OS) for the GBM patients were evaluated. Results Mean preoperative SCF plasma level of the GBM patients (2.80 ± 1.52 ng/ml) was significantly higher (p < 0.0001) than the healthy controls (0.80 ± 0.24 ng/ml) and patients with nonglial tumor (1.41 ± 0.76 ng/ml). Receiver operating characteristic analysis revealed that the preoperative SCF plasma level could distinguish the GBM patients from healthy controls and patients with nonglial tumors with the area under curve values of 0.915 and 0.790, respectively. However, no significant association was observed between the GBM patients’ preoperative SCF plasma levels and tumors’ volume (Spearman Rho correlation coefficient, 0.1847; 95% CI, p = 0.1652). The GBM patients were divided into two subgroups based on mean preoperative SCF plasma levels (2.80 ng/ml). No significant difference was observed between the patients’ PFS (p = 0.3792) and OS (p = 0.1469) at these two subgroups. Conclusion Taking together, the SCF plasma level can serve as a novel diagnostic blood-circulating biomarker for patients with GBM. However, its plasma level is not correlated with GBM patients’ tumor volume, PFS, or OS.
... Our findings are not consistent with observations from in vitro studies of pancreatic adenocarcinoma cell lines that demonstrated a tumoricidal effect of digoxin and other cardiac glycosides. One study reported induction of apoptosis at physiologically relevant levels of digoxin [11], while another reported suppression of HIF-1 [35,36] Should our observations prove to be correct, it would indicate that digoxin interacts with tumor pathogenesis through mechanisms that have yet to be described. Finally, our observation that digoxin exposure was not associated with survival are consistent with one small phase II clinical trial of pancreatic cancer patients in which Huachansu, a cardiac glycoside cousin of digoxin, did not increase or decrease survival when combined with chemotherapy [37]. ...
Article
Full-text available
Purpose Digoxin affects several cellular pathways involved in tumorigenesis. We sought to determine the association between digoxin use and pancreatic cancer risk and survival. Methods A nested case–control study using The Health Improvement Network (THIN), a population-representative database from the United Kingdom (UK). Cases included all individuals with incident diagnosis of pancreatic cancer. Each case was matched to up to four controls using incidence density sampling based on age, sex, practice site, calendar time, and duration of follow-up. Exposure of interest was digoxin therapy before cancer diagnosis. Odds ratios (ORs) and 95% confidence intervals (CIs) for the association between digoxin use and pancreatic cancer risk were estimated using conditional logistic regression. We further conducted a retrospective cohort study among pancreatic cancer cases using Cox regression model in order to evaluate the association between digoxin use and overall survival. Results We identified 4,113 cases with incident pancreatic cancer and 16,072 matched controls. The adjusted OR for diagnosis of pancreatic cancer among active digoxin users was 1.41 (95% CI 1.16–1.72). The risk did not change among active users with duration of therapy of more than 1 year (adjusted OR of 1.39, 95% CI 1.11–1.76). Digoxin was not associated with change in overall survival with an adjusted hazard ratio of 0.97 (95% CI 0.81–1.18). Conclusions Digoxin use was associated with modestly increased pancreatic cancer risk but did not affect overall survival.
Article
Resistance to chemotherapy in cancer leads to poor therapeutic outcomes and also leads to challenges in treatment. The present work evaluated the mechanism involved in the resistance of 5‐flurouracil (5‐FU) in pancreatic cancer. At least 14 different pancreatic cancer (PC) cell lines were used for the study. For in vivo study female nude mice were selected. Patient‐derived tumor xenograft samples were obtained from patients. The study involved, study for glucose uptake, fluorescence‐activated cell sorting for glucose transporter, 3‐[4,5‐dimethylthiazol‐2‐yl]‐2,5‐diphenyltetrazolium bromide for cell survival, Picto‐micrography for clonogenic assay, glutamine uptake assay, extracellular acidification and oxygen consumption rate, carbon dioxide release assay and lactate assay were also done. In addition to this, quantitative real‐time polymerase chain reaction analysis for expression of genes, chromatin immunoprecipitation assay, western blot for protein expression, and immunohistochemical analysis in tumor sections, the tumors were studied by imaging for hypoxia and localization of TKT and CTPS‐2. Also, patient‐derived xenograft tumors were engrafted in nude mice, followed by a glucose uptake assay. We reported that elevated glycolytic flux causes dependence on glucose in cancer cells and, at the same time, increases pyrimidine biosynthesis. It was also found that stem cell factor‐mediated stabilization of hypoxia‐inducible factor‐1a (HIF‐1α) modulates the resistance in PC. Targeting HIF‐1α in combination with 5‐FU, strongly reduced the tumor burden. The study concludes that stem cell factor modulates HIF‐1α and decreases the sensitivity in 5‐FU resistant pancreatic cancer cells by targeting glucose metabolism. Deceased expression levels of CTPS‐2 and TKT, which are regulators of pyrimidine biosynthesis could better the chance of survival in patients of pancreatic cancer receiving treatment of 5‐FU.
Article
Aims To investigate the regulatory mechanism of SCF expression in human GCs of PCOS related follicles. Materials and Methods SCF, BMP15 and HIF-1α were evaluated in human serums, follicular fluids (FFs) and GCs, which were collected from 69 PCOS patients and 74 normal ovulatory patients. KGN cell line was used in this study. Results Our results showed that the rate of MII oocyte and 2PN fertilization was lower in PCOS group, though PCOS patients retrieved much more oocytes. The level of BMP15 in FF and the level of SCF in serum and FF were also lower in PCOS patients. We found a weakened expression of HIF-1α and SCF in GCs from PCOS patients when compared with the non-PCOS patients. The expression of HIF-1α and SCF was significantly increased in KGN cells after treating cells with rhBMP15, however, this promotion effects of BMP15 on HIF-1α and SCF expression were obviously abolished by co-treatment with BMP-I receptor inhibitor (DM). Moreover, knock down of HIF-1α expression in KGN cells significantly reduced the expression of SCF in human GCs, in spite of activating BMP15 signaling pathway. Conclusions The present study suggest that BMP15 could induce SCF expression by up-regulating HIF-1α expression in human GCs, the aberrance of this signaling pathway might be involved in the PCOS related abnormal follicular development.
Article
Objectives: This study aimed to investigate the effect and mechanism of hypoxia on pancreatic cancer (PC) cell dedifferentiation and tumorigenic potential. Methods: Inhibition of hypoxia-inducible factor 1α (HIF-1α) and overexpression of Notch1 in PC HS766T cell lines were by lentiviral transfection. The expression of stem cell-specific markers C-X-C motif chemokine receptor 4, CD44, and Nestin was detected by immunofluorescence and Western blot assays. Cell invasion capacity was examined by Transwell assay. Tumorigenic potential was measured in an in situ tumor transplantation experiment. The expression of HIF-1α, Notch signals, and apoptosis signals was examined by Western blot assay. Results: Hypoxia promoted PC cells to dedifferentiate into stem-like cells by upregulating HIF-1α and activating Notch signals. Silencing of HIF-1α significantly repressed cell dedifferentiation and invasion, whereas overexpression of Notch1 reversed the effect of HIF-1α repression. In situ tumor transplantation experiment further confirmed that hypoxia promoted tumorigenic ability through upregulating HIF-1α. Moreover, the expression of HIF-1α and Notch1 was significantly increased in human PC tissues, and high expression of HIF-1α was correlated with poor survival rate. Conclusions: Hypoxia promoted PC cell dedifferentiation to stem-like cell phenotypes with high tumorigenic potential by activating HIF-1α/Notch signaling pathway, indicating a novel role in regulating PC progression.
Article
Full-text available
Stem cell factor (SCF), a ligand of c-kit, is a hematopoietic growth factor. Uncontrolled activity of SCF/c-kit signaling pathway contributes to the formation of a variety of human malignancies. In this study, we determined whether SCF expression could risk-stratify patients with hepatocellular carcinoma (HCC) after curative resection. HCC tissues from 160 patients were collected during curative resection and stained with SCF and CD34, a marker for microvessel density (MVD), using immunohistochemistry. Two statistical analyses were performed: an independent continuous and a multivariate categorical analysis, with test/validation set-defined cut points, and Kaplan-Meier estimated outcome measures of overall survival (OS) and relapse-free survival (RFS). We found that higher levels of SCF confer worse OS (continuous P = 0.014; and categorical P = 0.009), and RFS (continuous P = 0.002; categorical P = 0.003) of patients with HCC. SCF varies independently from MVD-CD34, tumor node metastasis, histologic grade, age and gender, and retains prognostic significance when analysed as a categorical variable in a multivariate analysis . We confirmed that MVD-CD34 is also an independent prognostic marker for patients with HCC. The levels of SCF and CD34 showed a positive and significant correlation (P < 0.0001) and double low expression confers superior OS (median = 48 months) and RFS (median = 24 months), whereas double high expression confers shortest RFS (median = 10.5 months) compared with single measurements. The prognostic values of SCF and CD34 were independently determined in this study and we propose that both of them are independent prognostic markers for HCC. © The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected] /* */
Article
Full-text available
Objective: Pancreatic ductal adenocarcinoma is a deadly disease because of late diagnosis and chemoresistance. We aimed to find a panel of serum cytokines representing diagnostic and predictive biomarkers for pancreatic cancer. Methods: A cytokine antibody array was performed to simultaneously identify 507 cytokines in sera of patients with pancreatic cancer and healthy controls. The nonparametric Mann-Whitney U test was used to pairwise compare the controls, the pretreated patients, and the posttreated patients. Fold changes greater than or equal to 1.5 or less than or equal to 1/1.5 were considered significant. Receiver operating characteristic curves were used to assess the performance of the model. A leave-one-out cross-validation was used for estimating prediction error. Results: Comparing the sera of pretreated patients against the control samples, the cytokines fibroblast growth factor 10 (FGF-10/keratinocyte growth factor-2 (KGF-2), chemokine (C-X-C motif) ligand 11 interferon inducible T cell alpha chemokine (I-TAC)/chemokine [C-X-C motif] ligand 11 (CXCL11), oncostatin M (OSM), osteoactivin/glycoprotein nonmetastatic melanoma protein B, and stem cell factor (SCF) were found significantly overexpressed. Besides, the cytokines CD30 ligand/tumor necrosis factor superfamily, member 8 (TNFSF8), chordin-like 2, FGF-10/KGF-2, growth/differentiation factor 15, I-TAC/CXCL11, OSM, and SCF were differentially expressed in response to treatment. Conclusions: We propose a role for FGF-10/KGF-2, I-TAC/CXCL11, OSM, osteoactivin/glycoprotein nonmetastatic melanoma protein B, and SCF as novel diagnostic biomarkers. CD30 ligand/TNFSF8, chordin-like 2, FGF-10/KGF-2, growth/differentiation factor 15, I-TAC/CXCL11, OSM, and SCF might represent as predictive biomarkers for gemcitabine and erlotinib response of patients with pancreatic cancer.
Article
Full-text available
There are at least four main signal transduction pathways within human cells which are activated by interaction of an extracellular ligand with its corresponding receptors. One of them is activation of protein kinase. Actually, any of these proteins at any level of the signaling cascade in a human cell can undergo mutation and cause irregular cellular proliferation and finally result in cancer. C-kit is alternatively called stem cell factor receptor (SCFR) or CD117. It appears that lack of c-kit expression accompanies progression of some tumors, e.g. lung, breast, GIST. The aim of this study was to evaluate C-kit protein expression level within cancer cases. Sixty specimens of breast cancer and 60 non-cancerous breast tissue specimens were evaluated by IHC for C-kit presentation. We used positive GIST slides as controls. Epi-info ver 6.04 (CDC, WHO) was used for analysis. C-kit was negative in all breast cancer specimens. C-kit was negative in 47 (78%) of 60 non-cancerous breast tissue specimens, but was positive in 13 (22%) of them (p < 0.0001). There is a reduction in C-kit expression with malignant transformation of breast epithelium. C-kit is believed to play a role in breast carcinogenesis. However, we should follow patients with normal or benign breast tissue to indicate any correlation between C-kit presentation and breast cancer development.
Article
Full-text available
Metastasis involves critical interactions between cancer and stromal cells. Intratumoral hypoxia promotes metastasis through activation of hypoxia-inducible factors (HIFs). We demonstrate that HIFs mediate para-crine signaling between breast cancer cells (BCCs) and mesenchymal stem cells (MSCs) to promote metastasis. In a mouse orthotopic implantation model, MSCs were recruited to primary breast tumors and promoted BCC metastasis to LNs and lungs in a HIF-dependent manner. Coculture of MSCs with BCCs augmented HIF activity in BCCs. Additionally, coculture induced expression of the chemokine CXCL10 in MSCs and the cognate receptor CXCR3 in BCCs, which was augmented by hypoxia. CXCR3 expression was blocked in cocultures treated with neutralizing antibody against CXCL10. Conversely, CXCL10 expression was blocked in MSCs cocultured with BCCs that did not express CXCR3 or HIFs. MSC coculture did not enhance the metas-tasis of HIF-deficient BCCs. BCCs and MSCs expressed placental growth factor (PGF) and its cognate receptor VEGFR1, respectively, in a HIF-dependent manner, and CXCL10 expression by MSCs was dependent on PGF expression by BCCs. PGF promoted metastasis of BCCs and also facilitated homing of MSCs to tumors. Thus, HIFs mediate complex and bidirectional paracrine signaling between BCCs and MSCs that stimulates breast cancer metastasis.
Article
Each year the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data were collected by the National Cancer Institute (Surveillance, Epidemiology, and End Results [SEER] Program), the Centers for Disease Control and Prevention (National Program of Cancer Registries), and the North American Association of Central Cancer Registries. Mortality data were collected by the National Center for Health Statistics. A total of 1,658,370 new cancer cases and 589,430 cancer deaths are projected to occur in the United States in 2015. During the most recent 5 years for which there are data (2007-2011), delay-adjusted cancer incidence rates (13 oldest SEER registries) declined by 1.8% per year in men and were stable in women, while cancer death rates nationwide decreased by 1.8% per year in men and by 1.4% per year in women. The overall cancer death rate decreased from 215.1 (per 100,000 population) in 1991 to 168.7 in 2011, a total relative decline of 22%. However, the magnitude of the decline varied by state, and was generally lowest in the South (15%) and highest in the Northeast (20%). For example, there were declines of 25% to 30% in Maryland, New Jersey, Massachusetts, New York, and Delaware, which collectively averted 29,000 cancer deaths in 2011 as a result of this progress. Further gains can be accelerated by applying existing cancer control knowledge across all segments of the population. CA Cancer J Clin 2015;000:000000. V C 2015 American Cancer Society.
Article
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers because of late symptoms and resistance to chemotherapy and radiation therapy. We have investigated the appearance of c-kit, a stem cell marker, in both normal adult pancreatic tissue and in cancerous tissue. Apart from some very pale staining of islets of Langerhans, normal pancreas was devoid of staining with antibodies to c-kit. In contrast, in cancerous tissue that still preserves the overall integrity of the pancreatic tissue, there was a clear labeling in islets of Langerhans, which seemed to be co-localized with insulin containing β cells. In other cases, where the pancreatic tissue was completely deteriorated, intensive labeling was clearly evident in remnants of both the exocrine and the endocrine tissues. The duct cells of the adenocarcinoma were moderately but clearly labeled with antibodies to c-kit. In contrast, in metastasis of PDAC, very intensive labeling of c-kit was evident. The location of KRAS, which is strongly associated with PDAC, was also analyzed at the initial stages of the disease, when islets of Langerhans still preserve their integrity to a large extent. KRAS was found exclusively in islets of Langerhans and overlapped in its location with insulin and c-kit expressing cells. It is suggested that the modulation of the expression of c-kit, visualized by antibodies to the oncogene molecule, may play an important role in the formation and progression of PDAC. The absence of c-kit in normal pancreas and its appearance in PDAC is probably due to a mutational event, which probably allows conversion of the β cells into cancer stem cells (CSC). Co-expression of both c-kit and KRAS, typical markers for CSC with overlapping with insulin in islets of Langerhans, strongly support the notion that β-cells play a central role in the development of PDAC. The use of specific drugs that can attenuate the kinase activity of c-kit or target KRAS expressing cancer cells should be tested in order to attenuate the progression of this lethal disease.
Article
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths expected in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival based on incidence data from the National Cancer Institute, the Centers for Disease Control and Prevention, and the North American Association of Central Cancer Registries and mortality data from the National Center for Health Statistics. A total of 1,660,290 new cancer cases and 580,350 cancer deaths are projected to occur in the United States in 2013. During the most recent 5 years for which there are data (2005-2009), delay-adjusted cancer incidence rates declined slightly in men (by 0.6% per year) and were stable in women, while cancer death rates decreased by 1.8% per year in men and by 1.5% per year in women. Overall, cancer death rates have declined 20% from their peak in 1991 (215.1 per 100,000 population) to 2009 (173.1 per 100,000 population). Death rates continue to decline for all 4 major cancer sites (lung, colorectum, breast, and prostate). Over the past 10 years of data (2000-2009), the largest annual declines in death rates were for chronic myeloid leukemia (8.4%), cancers of the stomach (3.1%) and colorectum (3.0%), and non-Hodgkin lymphoma (3.0%). The reduction in overall cancer death rates since 1990 in men and 1991 in women translates to the avoidance of approximately 1.18 million deaths from cancer, with 152,900 of these deaths averted in 2009 alone. Further progress can be accelerated by applying existing cancer control knowledge across all segments of the population, with an emphasis on those groups in the lowest socioeconomic bracket and other underserved populations. CA Cancer J Clin 2013;. © 2013 American Cancer Society.