ArticlePDF Available

Abstract and Figures

Purpose: The aim of this study was to evaluate the anti-angiogenic properties of soy isoflavones using two breast cancer cell lines, by measuring the concentration of 30 cytokines involved in angiogenesis using a multiplex glass slide ELISA-based array. Methods: Estrogen-dependent MCF-7 cells and estrogen-independent MDA-MB-231 cells were exposed to genistein (Gen), daidzein (Dai) and a soy seed extract (Ext) for 72 hrs, at selected concentration levels. The conditioned medium was analyzed using a glass slide, multiplex sandwich ELISA-based platform with fluorescent detection which allowed the identification and the quantification of 30 angiogenesis-related cytokines. Results: In MCF-7 cells, low, stimulatory concentrations of test compounds determined the increase of CXCL16 and VEGF-A level. Gen induced the greatest effect, with 1.5-fold change compared to control. When MDA-MB-231 cells were exposed to inhibitory concentrations, all test compounds determined a reduction of CXCL16 and VEGF-A level with approximately 30%. Conclusions: Soluble CXCL16 and VEGF-A are two promoters of angiogenesis and metastasis in breast cancer. The stimulation of these two angiogenesis-related cytokines could represent one of the mechanisms explaining the proliferative effects of low isoflavone doses in estrogen-dependent cells. In estrogen-independent cells, soy isoflavones inhibited their secretion, demonstrating promising anti-angiogenic properties.
Content may be subject to copyright.
JBUON 2018; 23 (Suppl 1): S53-S59
ISSN: 1107-0625, online ISSN: 2241-6293 • www.jbuon.com
E-mail: editorial_oce@jbuon.com
ORIGINAL ARTICLE
Correspondence to: Cristina Adela Iuga, PhD. Department of Proteomics and Metabolomics, MedFuture Research Center for
Advanced Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania.
Tel: +40-722-460-298, E-mail: iugac@umfcluj.ro
Received: 21/05/2018; Accepted: 16/06/2018
Inuence of soy isoavones in breast cancer angiogenesis: a
multiplex glass ELISA approach
Alina Uifalean1,2, Hermann Rath3, Elke Hammer3, Corina Ionescu4, Cristina Adela Iuga1,5,
Michael Lalk2
1
Department of Pharmaceutical Analysis, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy,
Cluj-Napoca, Romania; 2Institute of Biochemistry, University of Greifswald, Greifswald, Germany; 3Department of Functional
Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany;
4Department of Pharmaceutical Biochemistry and Clinical Laboratory, Faculty of Pharmacy, “Iuliu Haţieganu” University of
Medicine and Pharmacy, Cluj-Napoca, Romania;
5
Department of Proteomics and Metabolomics, MedFuture Research Center for
Advanced Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
Summary
Purpose: The aim of this study was to evaluate the anti-
angiogenic properties of soy isoavones using two breast
cancer cell lines, by measuring the concentration of 30 cy-
tokines involved in angiogenesis using a multiplex glass slide
ELISA-based array.
Methods: Estrogen-dependent MCF-7 cells and estrogen-
independent MDA-MB-231 cells were exposed to genistein
(Gen), daidzein (Dai) and a soy seed extract (Ext) for 72 hrs,
at selected concentration levels. The conditioned medium was
analyzed using a glass slide, multiplex sandwich ELISA-
based platform with uorescent detection which allowed the
identication and the quantication of 30 angiogenesis-
related cytokines.
Results: In MCF-7 cells, low, stimulatory concentrations
of test compounds determined the increase of CXCL16 and
VEGF-A level. Gen induced the greatest eect, with 1.5-fold
change compared to control. When MDA-MB-231 cells were
exposed to inhibitory concentrations, all test compounds de-
termined a reduction of CXCL16 and VEGF-A level with ap-
proximately 30%.
Conclusions: Soluble CXCL16 and VEGF-A are two pro-
moters of angiogenesis and metastasis in breast cancer.
The stimulation of these two angiogenesis-related cytokines
could represent one of the mechanisms explaining the prolif-
erative eects of low isoavone doses in estrogen-dependent
cells. In estrogen-independent cells, soy isoavones inhibited
their secretion, demonstrating promising anti-angiogenic
properties.
Key words: angiogenesis, breast cancer cells, CXCL16, ELI-
SA, isoavones, VEGF-A
Introduction
In the USA, breast, lung and colorectal cancers
account for 50% of all cancer cases expected to oc-
cur in women in 2018. Of this percentage, breast
cancer alone accounts for 30%, which embody
266,120 new diagnosed cases [1]. Breast cancer
rates are generally higher in Northern America,
Australia/New Zealand, Western Europe and low
in most of Africa and Asia [2].
Several studies have related the low incidence
rates of breast cancer in Asian countries with the lo-
cal dietary patterns, showing that soy consumption
could lower the risk of breast cancer for both pre-
and post-menopausal women in Asian countries
[3,4]. Later, these epidemiological observations
were strengthened by in vitro data, soy isoavones
and especially genistein, showing antiproliferative
This work by JBUON is licensed under a Creative Commons Attribution 4.0 International License.
Soy isoavones in breast cancer angiogenesis54
JBUON 2018; 23 (Suppl 1): S54
eects by sustaining apoptosis, antioxidant defense
and DNA repair and, not least, by inhibiting the
development of tumor angiogenesis and metastasis
[5].
Soy isoavones have been explored as prom-
ising anti-angiogenetic agents as they appear to
inhibit multiple angiogenic mechanisms, such as
regulation of vascular endothelial growth factor
(VEGF), matrix metalloproteinases (MMPs), epi-
dermal growth factor receptor (EGFR) expressions
and NF-κB, PI3-K/Akt or ERK1/2 signaling path-
ways [5,6]. However, despite the intensive research,
the anti-angiogenic potential of soy isoavones in
breast cancer remains controversial, mainly due to
their twofold eect [7].
Several in vitro assays have been developed
to assess the angiogenic properties of exogenous
agents. Most models focus on proliferation, mi-
gration, and dierentiation of endothelial cells
[8]. While these tests determine the eect or the
outcome of drugs on blood vessel formation, more
high-throughput tests have been developed in order
to identify which particular angiogenic molecules
or mechanisms are targeted by the test compounds.
Such are the glass slide ELISA-based quantitative
systems, extensively used for the rapid proling of
cytokine expression.
The main advantage of glass slides over the
single-targeted 96-well plate ELISAs or Western
blots is the possibility of performing simultane-
ous identication and quantication of multiple
cytokines, growth factors, proteases, soluble recep-
tors and other angiogenesis-associated proteins in
a single experiment. Furthermore, they are highly
specic and reproducible, require low sample vol-
umes and are well-suited for high throughput as-
says [9].
To our knowledge, no ELISA-based quantitative
array on breast cancer cells exposed to isoavones
has been performed so far. The identication and
quantication of the key molecules involved in an-
giogenesis will provide a further understanding of
isoavones’ anti-angiogenic properties. The aim of
this study was to evaluate the anti-angiogenic prop-
erties of genistein (Gen), daidzein (Dai) and a soy
seed extract (Ext) using two breast cancer cell lines,
MCF-7 and MDA-MB-231, by measuring the con-
centration of 30 cytokines involved in angiogenesis
using a multiplex glass slide ELISA-based array.
Methods
Chemical and standards
All chemicals and standards were purchased from
Sigma-Aldrich (Taufkirchen, Germany), unless otherwise
stated.
The soy extract was purchased from Hunan Gold-
liloo Pharmaceutical Co., Ltd. (Changsha, China). Accord-
ing to manufacturer’s specications, the extract was ob-
tained from soy seeds (Glycine max), using an aqueous
ethanolic solution followed by spray-drying. The extract
contains 40% isoavones, of which daidzein represents
only 1.50%, glycitein 0.12%, and genistein 0.02%. The
isoavone distribution was conrmed in our laboratory
by a validated HPLC-UV method [10].
Stock solutions of standard Gen, Dai, and Ext were pre-
pared in dimethyl sulfoxide (DMSO) and stored at -20°C.
Cell culture and culture conditions
The MCF-7 and MDA-MB-231 breast adenocarci-
noma cell lines were obtained from CLS Cell Lines Ser-
vice (Eppelheim, Germany) and routinely cultured as
previously described [11]. All cells used in experiments
were between passage number 5 and 20.
Cell treatment and sampling
The test concentrations of Gen, Dai, and Ext were
established based on a MTT test, as previously described
[11]. Briey, in MCF-7 estrogen-dependent cells, all com-
pounds induced a twofold eect, stimulating cell growth
at relatively low concentrations and causing inhibition
at higher concentrations. Therefore, we selected two con-
centration levels for each test compound: the concentra-
tions that stimulated cell proliferation by 20% compared
to control (SC
20
) and the concentrations that inhibited
cell growth by 20% compared to control (IC20). The SC20
concentrations for Gen, Dai, and Ext were 5.62 μM, 19.01
μM, and 22.59 μg/mL respectively, while the IC20 con-
centrations were 22.44 μM, 52.24 μM, and 166.34 μg/mL
respectively. For MDA-MB-231 estrogen-independent
cells, only a dose dependent inhibitory eect was ob-
served and, therefore, only the IC20 concentrations were
selected. Precisely, these IC
20
concentrations were 11.04
μM for Gen, 36.39 μM for Dai, and 26.36 μg/mL for Ext
[11].
For cell treatment, 2.4×106 MCF-7 cells or 1.2×106
MDA-MB-231 cells were seeded in 150 mm cell cul-
ture dishes (Sarstedt, Germany) in 15 mL RPMI 1640
medium supplemented with 10% heat-inactivated fetal
bovine serum, 1 mM sodium pyruvate, 1% non-essential
amino acids and 1% penicillin-streptomycin.
The dishes were shaken for 1 min to ensure the
homogeneous distribution of cells. Next, all plates were
incubated for 24 hrs to allow cell attachment. Aer 24
hrs, the medium was replaced with 28 mL fresh medium
containing the selected concentrations of Gen, Dai, Ext,
or DMSO as solvent control. In all cases, the nal con-
centration of DMSO did not exceed 0.01%. The incuba-
tion time was 72 hrs.
For sampling, 1.5 mL conditioned medium were cen-
trifuged at 2000 rpm, at 4°C for 10 min. The supernatant
was immediately frozen at -80°C until measurement.
Antibody array analysis of angiogenesis related cytokines
For the quantication of angiogenesis-associated
cytokines, we used Quantibody Human Angiogenesis
Array 3 (#QAH-ANG-3, RayBiotech, Norcross, Georgia,
Soy isoavones in breast cancer angiogenesis 55
JBUON 2018; 23 (Suppl 1): S55
USA). This is a glass slide, multiplex sandwich ELISA-
based platform which allows the identication and quan-
tication of 30 cytokines, chemokines, growth factors,
and other molecules involved in angiogenesis. The 30
spotted targets were angiogenin-1, angiostatin, C-X-C
motif chemokine ligand 16 (CXCL16), epidermal growth
factor, broblast growth factor 4, follistatin, granulo-
cyte colony-stimulating factor, granulocyte-macrophage
colony-stimulating factor, I-309, interleukin-1 beta, in-
terleukin-4, interleukin-10, interleukin-12 subunit p40,
interleukin-12 subunit p70, interferon-inducible T-cell
alpha chemoattractant, monocyte chemotactic protein 2,
monocyte chemotactic protein 3, monocyte chemotactic
protein 4, matrix metalloproteinase-1, matrix metallo-
proteinase-9, platelet endothelial cell adhesion mole-
cule-1, transforming growth factor alpha, transforming
growth factor beta-3, tyrosine-protein kinase receptor
Tie-1, tyrosine-protein kinase receptor Tie-2, urokinase
plasminogen activator surface receptor, vascular en-
dothelial growth factor-A (VEGF-A), vascular endothelial
growth factor receptor 2, vascular endothelial growth
factor receptor 3 and vascular endothelial growth factor
D. Each antibody, together with two positive controls and
a negative control, is printed in four identical spots, so
each cytokine is measured four times per sample.
The assay was conducted according to manufacturer
recommended protocol [9]. Briey, the glass slides were
rst allowed to equilibrate and dry at room temperature
for 2 hrs. In the blocking step, 100 μL sample diluent was
added into each well and the slides were incubated for
30 min at room temperature. Next, the sample diluent
was discarded and 100 μL calibration standard cytokines
or conditioned medium were added into each well. The
glass chamber was covered with adhesive lm and incu-
bated overnight, at 4°C, on a plate shaker (Titramax 101,
Heidolph Instruments, Schwabach, Germany) at 200 rpm.
On the next day, the supernatant was discarded and
each well was washed ve times with Wash Buer I
and two times with Wash Buer II. Subsequently, the
biotinylated antibody cocktail was reconstituted and
80 μL were added per well. Aer 2 hrs, the antibody
cocktail was removed, the wells were washed again with
the two washing buers and 80 μL of Cy3 Equivalent
Dye-Streptavidin were added per well. The slides were
incubated in the dark, at room temperature for 1 hr. Aer
other washing steps, the slides were carefully removed
from the gasket and allowed to dry at room temperature.
For uorescence detection, a DNA Microarray Scan-
ner (G2505C, Agilent Technologies, USA) with a scan
resolution of 10 μm was used.
Data analysis
For background subtraction and densitometry
measurement, the scanned images were analyzed using
Image Studio Lite (v.2.5.2.). For each spot, the dened
area for signal capture was a circle with a 158-micron
diameter. The median intensity of a three-pixel border
around the dened circle was used for local background
subtraction.
As our cells were cultivated in serum-containing
medium, which might contain various types of cytokines,
the median signal intensity of each cytokine of the com-
plete medium array was subtracted from the signal in-
tensity of the corresponding cytokine from each other
array.
Next, data normalization was carried out by ac-
counting for the dierences in signal intensities of
the positive control spots across all arrays. The posi-
tive control spots represent standardized amounts of
biotinylated antibody and the signal of these spots is
dependent on the amount of streptavidin-uor bound
to that antibody. This bounding capacity will propor-
tionally aect the signal intensity of every spot on the
array. Therefore, the dierences in the positive control
signals between arrays will accurately reect the dier-
ences between other spots on those arrays. The reference
array was the solvent control (medium with DMSO only)
corresponding to each cell line. The normalized values
were calculated using equation 1 [9]:
nX(Y)=X(Y)×P / P(Y)
nX(Y)= the normalized value for cytokine “X” of sample
“Y”, X(Y)= the signal density of the spots for cytokine
“X” of sample “Y”, P= the average signal density of the
positive control spots on the reference array, P(Y)= the
average signal density of the positive control spots of
sample “Y”
Statistics and visualization
The calibration curves and the statistical analysis
were executed using Prism (v.6.01, GraphPad Soware).
Each treatment was compared to the corresponding sol-
vent control according to two-way ANOVA with Sidak’s
pg/mL Fold change
CXCL16 concentration
In MCF-7 cells
Control 1140.91
Gen SC20 1775.65 1.55
Dai SC20 1691.43 1.48
Dai IC20 547.37 -2.08
In MDA-MB-231 cells
Control 1574.22
Gen IC20 1232.29 -1.28
Ext IC20 1137.21 -1.38
VEGF-A concentration
In MCF-7 cells
Control 1309.22
Gen SC20 1982.84 -1.51
Ext SC20 1808.55 -1.38
In MDA-MB-231 cells
Control 1521.43
Gen IC20 1192.17 -1.27
Dai IC20 1130.65 -1.34
Ext IC20 950.75 -1.60
Table 1. The concentration and the fold change of
signicantly altered cytokines (p<0.05, two-way ANOVA
with Sidak’s correction for multiple comparisons)
Soy isoavones in breast cancer angiogenesis56
JBUON 2018; 23 (Suppl 1): S56
correction for multiple comparisons. Dierences with
p values less than 0.05 were considered as statistically
signicant.
Results
Exposure of both breast cancer cell lines to
Gen, Dai, and Ext induced signicant changes, es-
pecially in the signal intensity of two cytokines,
CXCL16 and VEGF-A. In MCF-7 cells, SC20 of test
compounds caused an increase in CXCL16 and
VEGF-A signal intensity, while treatment of cells
with IC20 concentrations led to a reduction of
CXCL16 level. For MDA-MB-231 cells, inhibitory
concentrations of test compounds triggered a de-
crease in the VEGF-A and CXCL16 signal intensity
(Figure 1).
Next, the mean intensity of the signicantly
changed cytokines was plotted on the correspond-
ing calibration curve (Figure 2). Using these curves,
the absolute cytokine concentration was then cal-
culated (Table 1).
Discussion
Soy isoavones are known as promising anti-
angiogenic agents, acting on multiple pathways,
such as ERK1/2 signaling pathway, regulation of
Figure 1. The signal intensity of CXCL16 and VEGF-A aer MCF-7 and MDA-MB-231 cells were exposed to genistein
(Gen), daidzein (Dai), and soy extract (Ext) at test concentrations. Asterisks indicate statistically signicant dierences
(p<0.05) between solvent control and treated samples.
Figure 2. The calibration curves for CXCL16 and VEGF-A obtained by plotting their mean intensities against the pre-
determined concentrations. The curves were generated using a non-linear regression t model (R2=0.9974 for CXCL16
and R2=0.9584 for VEGF-A).
Soy isoavones in breast cancer angiogenesis 57
JBUON 2018; 23 (Suppl 1): S57
VEGF or MMPs expression [5,6]. However, there is
limited data regarding the inuence of these natu-
ral compounds on CXCL16 expression.
CXCL16, along with CXCL12 (C-X-C motif
chemokine ligand 12), belong to the superfamily of
chemotactic cytokines, which govern the immune
cell tracking between or within tissues. Through
coordinated interaction with its specic receptor,
CXCR6 (C-X-C motif chemokine receptor 6), CXCL16
also plays a crucial role in tumor growth, invasion,
angiogenesis, and metastasis in various types of
cancers such as breast adenocarcinoma [12-14],
lung cancer [15] or prostate cancer [16]. Moreover,
for prostate and breast cancers, a positive correla-
tion between CXCR6/CXCL16 expression and cancer
aggressiveness was found [17,18], higher CXCR6 ex-
pression in nest site and metastatic lymph node be-
ing responsible for breast cancer progression [12].
So far, in vitro studies have shown that soy iso-
avones can regulate other angiogenic chemokines,
such as CXCL12. In MCF-7 estrogen-dependent
cells, low doses of Gen or Dai (1–10 μM) induced a
signicant increase in CXCL12 level [19-21], trig-
gering cell proliferation and invasion. When the
same cell line was exposed to higher Gen concen-
trations (>25 μM), the CXCL12 mRNA level was
signicantly downregulated. This downregulation
resulted in a subsequent inhibition of migration
and invasion. In MDA-MB-231 cells, CXCR4 (C-X-C
motif chemokine receptor 4), the cognate receptor
of CXCL12, was downregulated by Gen in a dose-
dependent manner [19].
To our knowledge, no study has assessed the ef-
fect of soy isoavones on CXCL16 chemokine so far.
Our results show that isoavone treatment
triggers similar changes for soluble CXCL16 ex-
pression, as for CXCL12. Low doses of isoavones
(SC
20
) signicantly stimulated CXCL16 secretion
in MCF-7 cells, Gen causing the highest CXCL16
increase. When MCF-7 were exposed to higher, in-
hibitory doses of isoavones (IC20), only Dai caused
a signicant decrease. The CXCL16 decrease caused
by Dai could be due to the anti-inammatory prop-
erties of Dai, which was shown to suppress the
transcription of pro-inammatory chemokines,
such as CXCL2, by depressing PARP-1 activity [22].
However, the IC20 of Gen used in this study
(22.44 μM for MCF-7 cells) was lower than the con-
centrations used in other studies [19,23]. Therefore,
it is not excluded that higher Gen concentrations,
most likely >50 μM, could decrease the CXCL16 se-
cretion. In MDA-MB-231 cells, all test compounds
generated a decrease in the CXCL16 level.
One of the mechanisms proposed for explain-
ing the proliferative eects of CXCR6/CXCL16
breast cancer cells involves the activation of down-
stream signaling paths, such as ERK1/2 signaling
pathway [12]. Apparently, stimulation of ERK1/2
pathway activates RhoA, a member of the RhoGT-
Pase family. The eect leads to inhibition of colin
activity, responsible for the regeneration of actin
laments. In response to colin inhibition, F-actin
stability enhances, favoring breast cancer invasive-
ness and metastasis [12]. In fact, Gen can also act
as direct modulator of ERK1/2 pathway, promoting
MCF-7 cell growth through delayed and prolonged
phosphorylation of ERK1/2 [24].
An alternative explanation could rely on the
estrogenic eects of low isoavones doses. Similar
to estrogen, which upregulates CXCL12 and CXCR4
expression in breast cancer cells [25], isoavones
could upregulate CXCL16 expression and secretion
acting through the same molecular mechanisms.
Isoavone treatment also determined signi-
cant changes in the VEGF-A level. Compared to
CXCL16, VEFG-A is a much more known player in
the process of tumor angiogenesis and the most
intensively studied member of VEGF family. Ac-
tivation of the VEGF-receptor pathway triggers a
network of signaling processes that promote cell
growth, migration, and survival from pre-existing
vasculature. The concentrations of the VEGF pro-
tein and VEGF receptors in the serum of breast
cancer patients showed positive correlations with
estrogen receptor status and the clinical stage of
disease [26].
Soy isoavones, and particularly Gen, have
been intensively examined for their potential to
modulate VEGF-A secretion, especially using cul-
tured human umbilical vein endothelial cells [6,27].
In breast cancer, low concentrations (10-12-10-6 M)
of Gen have similar eects as estrogen in estrogen
receptor (ER) positive cells like MCF-7 (ER posi-
tive), MELN (derived from MCF-7 cells) and MELP
(derived from MDA-MB-231 cells and transfected
with ER) inducing VEGF-A expression signicantly.
The same eect was not observed in MDA-MB-231
cells, suggesting that ER is necessary for VEGF
stimulation [28]. On the other hand, when MDA-
MB-453 cells were exposed to high Gen concen-
tration, the VEGF mRNA expression decreased
signicantly [6] pointing to a second receptor and
signaling pathway for Gen.
Our results are in line with the existing data,
that low, stimulatory concentrations of Gen or Ext
increase VEGF-A secretion in MCF-7 cells. Appar-
ently, low isoavone doses seem to mimic again
the estrogen action, stimulating the secretion of
VEGF-A. As VEGF-A promotes cell proliferation,
upregulation of VEGF-A secretion could be one of
the mechanisms explaining the proliferative eects
of isoavones.
Soy isoavones in breast cancer angiogenesis58
JBUON 2018; 23 (Suppl 1): S58
Notably, concentrations of Gen below 5 μM
correspond to a blood plasma concentration attain-
able in a soy-rich diet [29]. As CXCL16 and VEGF-A
secretion were both stimulated at low isoavone
concentrations, special attention should be paid
to the daily phytoestrogen intake by patients with
estrogen responsive breast cancer subtype to avoid
any pro-angiogenic eects.
In estrogen-independent MDA-MB-231 cells,
IC
20
of all test compounds triggered VEGF-A de-
crease. Inhibition of VEGF-A expression is a poten-
tial strategy especially in the triple negative breast
cancer, the cancer subtype that lacks any targeted
therapy and with the worst prognosis among all
breast cancer subtypes. As isoavones are capable
of inhibiting VEGF-A secretion in MDA-MB-231
cells, they could represent promising anti-angio-
genic agents.
Conclusion
Our study investigated the potential of soy iso-
avones to modulate the main molecules involved
in angiogenesis, using a quantitative glass slide
ELISA-based array. The results showed that iso-
avones exert dose dependent eects in both cell
lines: in MCF-7 cells, low isoavone doses stimu-
lated the secretion of CXCL16 and VEGF-A, two
promoters of angiogenesis and metastasis, while
higher concentrations inhibited CXCL16 and VEGF-
A secretion in MDA-MB-231 cells. The anti-angi-
ogenic properties of isoavones could be further
exploited as an eective strategy, especially in tri-
ple negative breast cancers.
Acknowledgements
This work was supported by the “Iuliu
Haţieganu” University of Medicine and Phar-
macy Cluj-Napoca through Internal Grant No.
1491/20/28.01.2014 and the People Programme
(Marie Curie Actions) of the European Union’s Sev-
enth Framework Programme FP7/2007-2013/under
REA Grant Agreement No. 317338.
We are grateful to Philipp Westho and Ra-
mona Suharoschi for their helpful discussions and
advice.
Conict of interests
The authors declare no conict of interests.
References
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018.
CA Cancer J Clin 2018;68:7-30.
2.
Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent
J, Jemal A. Global cancer statistics, 2012. CA Cancer J
Clin 2015;65:87-108.
3. Chen M, Rao Y, Zheng Y et al. Association between soy
isoavone intake and breast cancer risk for pre- and
post-menopausal women: a meta-analysis of epidemio-
logical studies. PLoS One 2014;9(2):e89288.
4.
Nagata C, Mizoue T, Tanaka K et al. Soy intake and
breast cancer risk: an evaluation based on a systematic
review of epidemiologic evidence among the Japanese
population. Jpn J Clin Oncol 2014;44:282-95.
5. Uifalean A, Schneider S, Ionescu C, Lalk M, Iuga CA.
Soy isoavones and breast cancer cell lines: molecu-
lar mechanisms and future perspectives. Molecules
2015;22:21.
6. Varinska L, Gal P, Mojzisova G, Mirossay L, Mojzis J.
Soy and breast cancer: focus on angiogenesis. Int J Mol
Sci 2015;16:11728-49.
7.
Allred CD, Allred KF, Ju YH, Virant SM, Helferich
WG. Soy diets containing varying amounts of gen-
istein stimulate growth of estrogen-dependent (MCF-
7) tumors in a dose-dependent manner. Cancer Res
2001;61:5045-50.
8. Tahergorabi Z, Khazaei M. A review on angiogenesis
and its assays. Iran J Basic Med Sci 2012;15:1110-26.
9. RayBiotech - Quantibody Human Angiogenesis Array
3 - User Manual [29.03.2018]. Available from: https://
www.raybiotech.com/quantibody-human-angiogene-
sis-array-3-1-slide/.
10.
Uifalean A, Farcas A, Ilies M, Heghes SC, Ionescu C,
Iuga CA. Assessment of isoavone aglycones variabil-
ity in soy food supplements using a validated HPLC-UV
method. Clujul Med 2015;88:373-80.
11. Uifalean A, Schneider S, Gierok P, Ionescu C, Iuga CA,
Lalk M. The impact of soy isoavones on MCF-7 and
MDA-MB-231 breast cancer cells using a global me-
tabolomic approach. Int J Mol Sci 2016;17:1443.
12.
Xiao G, Wang X, Wang J et al. CXCL16/CXCR6
chemokine signaling mediates breast cancer progres-
sion by pERK1/2-dependent mechanisms. Oncotarget
2015;6:14165-78.
13.
King J, Mir H, Singh S. Association of cytokines and
chemokines in pathogenesis of breast cancer. Prog Mol
Biol Transl Sci 2017;151:113-36.
14.
Fang Y, Henderson FC, Jr., Yi Q, Lei Q, Li Y, Chen N.
Chemokine CXCL16 expression suppresses migration
and invasiveness and induces apoptosis in breast can-
cer cells. Mediators Inamm 2014;2014:478641.
Soy isoavones in breast cancer angiogenesis 59
JBUON 2018; 23 (Suppl 1): S59
15. Liang K, Liu Y, Eer D, Liu J, Yang F, Hu K. High CXC
chemokine ligand 16 (CXCL16) expression promotes
proliferation and metastasis of lung cancer via reg-
ulating the NF-kappaB pathway. Med Sci Monit
2018;24:405-11.
16.
Richardsen E, Ness N, Melbo-Jorgensen C et al. The
prognostic signicance of CXCL16 and its receptor C-X-
C chemokine receptor 6 in prostate cancer. Am J Pathol
2015;185:2722-30.
17. Lu Y, Wang J, Xu Y et al. CXCL16 functions as a novel
chemotactic factor for prostate cancer cells in vitro.
Mol Cancer Res 2008;6:546-54.
18. Deng L, Chen N, Li Y, Zheng H, Lei Q. CXCR6/CXCL16
functions as a regulator in metastasis and progression
of cancer. Biochim Biophys Acta 2010;1806:42-9.
19.
Hsu EL, Chen N, Westbrook A et al. Modulation of
CXCR4, CXCL12, and tumor cell invasion potential in
vitro by phytochemicals. J Oncol 2009;2009:491985.
20. Habauzit D, Boudot A, Kerdivel G, Flouriot G, Pakdel F.
Development and validation of a test for environmental
estrogens: checking xeno-estrogen activity by CXCL12
secretion in breast cancer cell lines (CXCL-test). Envi-
ron Toxicol 2010;25:495-503.
21. Lecomte S, Lelong M, Bourgine G, Efstathiou T, Sali-
gaut C, Pakdel F. Assessment of the potential activity
of major dietary compounds as selective estrogen re-
ceptor modulators in two distinct cell models for pro-
liferation and dierentiation. Toxicol Appl Pharmacol
2017;325:61-70.
22.
Li HY, Pan L, Ke YS et al. Daidzein suppresses pro-
inflammatory chemokine Cxcl2 transcription in
TNF-alpha-stimulated murine lung epithelial cells
via depressing PARP-1 activity. Acta Pharmacol Sin
2014;35:496-503.
23. Lee WY, Huang SC, Tzeng CC, Chang TL, Hsu KF. Al-
terations of metastasis-related genes identied using
an oligonucleotide microarray of genistein-treated
HCC1395 breast cancer cells. Nutr Cancer 2007;58:239-
46.
24.
Liu H, Du J, Hu C et al. Delayed activation of extracellu-
lar-signal-regulated kinase 1/2 is involved in genistein-
and equol-induced cell proliferation and estrogen-re-
ceptor-alpha-mediated transcription in MCF-7 breast
cancer cells. J Nutr Biochem 2010;21:390-6.
25.
Boudot A, Kerdivel G, Habauzit D et al. Dierential
estrogen-regulation of CXCL12 chemokine recep-
tors, CXCR4 and CXCR7, contributes to the growth
eect of estrogens in breast cancer cells. PLoS One
2011;6(6):e20898.
26.
Thielemann A, Baszczuk A, Kopczynski Z, Kopczynski P,
Grodecka-Gazdecka S. Clinical usefulness of assessing
VEGF and soluble receptors sVEGFR-1 and sVEGFR-2
in women with breast cancer. Ann Agric Environ Med
2013;20:293-7.
27.
Yu X, Zhu J, Mi M, Chen W, Pan Q, Wei M. Anti-an-
giogenic genistein inhibits VEGF-induced endothelial
cell activation by decreasing PTK activity and MAPK
activation. Med Oncol 2012;29:349-57.
28. Buteau-Lozano H, Velasco G, Cristofari M, Balaguer P,
Perrot-Applanat M. Xenoestrogens modulate vascular
endothelial growth factor secretion in breast cancer
cells through an estrogen receptor-dependent mecha-
nism. J Endocrinol 2008;196:399-412.
29. Cassidy A, Brown JE, Hawdon A et al. Factors aecting
the bioavailability of soy isoavones in humans aer
ingestion of physiologically relevant levels from dif-
ferent soy foods. J Nutr 2006;136:45-51.
... Thus, it is reasonable that targeting the HIF-1α/HIF axis may represent a promising antiangiogenic and anticancer therapeutic approach [72]. Furthermore, in line with the action of the flavonoid explained above, by using a dedicated multiplex-array assay, Uifalean and co-workers (2018) showed genistein dosedependently hindered C-X-C motif chemokine ligand 16 (CXCL16) and vascular endothelial growth factor-A (VEGFA) secretion in BC in vitro (in MCF-7 estrogendependent and MDA-MB-231 estrogen-independent cell lines) [73]. Interestingly, both CXCL16 and VEGFA not only trigger angiogenesis but also promote metastasis [73,74]. ...
... Furthermore, in line with the action of the flavonoid explained above, by using a dedicated multiplex-array assay, Uifalean and co-workers (2018) showed genistein dosedependently hindered C-X-C motif chemokine ligand 16 (CXCL16) and vascular endothelial growth factor-A (VEGFA) secretion in BC in vitro (in MCF-7 estrogendependent and MDA-MB-231 estrogen-independent cell lines) [73]. Interestingly, both CXCL16 and VEGFA not only trigger angiogenesis but also promote metastasis [73,74]. More investigations reveal that genistein inhibits angiogenesis in other tumor types [75,76]. ...
Article
Full-text available
Genistein (4′,5,7-trihydroxyisoflavone) is a phytoestrogen belonging to a subclass of natural flavonoids that exhibits a wide range of pharmacological functions, including antioxidant and anti-inflammatory properties. These characteristics make genistein a valuable phytochemical compound for the prevention and/or treatment of cancer. Genistein effectively inhibits tumor growth and dissemination by modulating key cellular mechanisms. This includes the suppression of angiogenesis, the inhibition of epithelial–mesenchymal transition, and the regulation of cancer stem cell proliferation. These effects are mediated through pivotal signaling pathways such as JAK/STAT, PI3K/Akt/mTOR, MAPK/ERK, NF-κB, and Wnt/β-catenin. Moreover, genistein interferes with the function of specific cyclin/CDK complexes and modulates the activation of Bcl-2/Bax and caspases, playing a critical role in halting tumor cell division and promoting apoptosis. The aim of this review is to discuss in detail the key cellular and molecular mechanisms underlying the pleiotropic anticancer effects of this flavonoid.
... Notably, Genistein flavonoids exert inhibitory effects on angiogenesis in rheumatoid arthritis by targeting IL-6/JAK2/STAT3/VEGF signaling pathway 29 . Moreover, Genistein inhibited angiogenesis and inflammation in a mouse model of peritoneal endometriosis 20 and suppressed the expression of CXCL16 and VEGF-A in breast cancer cells 30 . In our animal experiments, the administration of Genistein demonstrated a significant decrease in peritoneal vessel density. ...
Article
Full-text available
Peritoneal fibrosis has been linked to hypoxia-inducible factor 1-alpha (HIF-1α) as well as O-linked-N-acetylglucosaminylation (O-GlcNAcylation) in peritoneal dialysis (PD). Genistein, recognized for its HIF-1α inhibitory and antifibrotic effects, presents a potential intervention against peritoneal mesothelial-mesenchymal transition (MMT) as well as fibrosis in PD. This study employed human peritoneal mesothelial cells (HPMCs) together with adenine-induced chronic kidney disease (CKD) rats undergoing peritoneal dialysis to explore Genistein’s role in high glucose-induced peritoneal MMT and fibrosis. Our findings reveal that Genistein exerts anti-MMT and anti-fibrotic effects by inhibiting HIF-1α in HPMCs under high glucose conditions. Genistein inhibited O-GlcNAcylation status of HIF-1α through the mTOR/O-GlcNAc transferase (OGT) pathway, promoting its ubiquitination as well as the subsequent proteasomal degradation. In adenine-induced CKD rats undergoing peritoneal dialysis, Genistein suppressed the mTOR/OGT expression and reduced the abundance of O-GlcNAcylation along with HIF-1α in the peritoneum. Additionally, Genistein protected against increased peritoneal thickness, fibrosis, and angiogenesis, while improving peritoneal function. Based on our results, it could be inferred that Genistein might inhibit the abundance of HIF-1α via the mTOR/OGT pathway, thereby ameliorating MMT as well as fibrosis in PD.
... In estrogen-independent cells, daidzein inhibited their secretion, indicating anti-angiogenic properties. Uifalean et al., 2018 The utilization of daidzein is associated with a reduced risk of prostate cancer but the direct effects of daidzein on the androgen receptor (AR) signaling pathway are not well understood. ...
Chapter
Currently, soybean (Glycine max (L.) Merrill), a Leguminosae family member, has become one of the main economical oilseed beans. It is being cultivated nowadays in all major areas of the world including China, Japan, Brazil, the USA, and Korea as well as in many South and Midwest countries for several uses. The reason lies in the introduction of multiple local varieties, efficient seed supply, and timely technology transfer, participation of the public sector as well as large international capital groups, and large-scale introduction of new soy foods. It is primarily being cultivated as a substitute for high-protein meat and a source of vegetable oil. Furthermore, the availability of many bioactive compounds has also increased the interest of various researchers toward this bean which originated from northeast China. As a result, it has emerged somewhere as one of the nutritious cum economical parts of the vegan diet. Due to its nutritive value, this “yellow meat of the field” is touted by many as a potential weapon against global hunger. Next to diet, soybean and related greater market value products are being employed either directly or as an ingredient in making cheese, spreads, paints, fertilizers, adhesives, fire extinguisher fluids, animal feed, etc. Due to all these applications, soybean was cultivated on nearly 125 million hectares of the area resulting in 348.7 million tons of harvest in the year 2018. This quantity of production is projected to increase in the near future with a parallel surge in purchasing demand of the every second increasing population. However, there are still many “yield limiters” that uneven the soybean production at both pilot and global scales by nearly 50%. In order to tackle all these soybean yield limiters in a highly efficient manner, various techniques including cross hybridization, molecular marker-assisted breeding, transgenic breeding, tilling, microbiome engineering, and genome editing are being employed by various research groups. Therefore, in the present chapter, the focus is solely on how with time the soybean has proved its strong candidature as a key player for global food security. Furthermore, the production trends at the world and Indian scale are also highlighted. Additionally, the present chapter is an attempt to provide a streamlined overview of all these soybean yield limiters and employed technologies, in brief, to pave the way for the readers for other chapters in the book.KeywordsSoybeanChinaNutritionYieldYield limiters
Article
Background Cancer metastasis is one of the major clinical challenges worldwide and is highly associated with patient's deaths. Therefore, targeting metastatic related proteins is the key to the development of anticancer therapy. Osteopontin (OPN) overexpression is associated with breast cancer progression and metastasis; thus, it is considered a promising biomarker. Many in vitro and in vivo studies have been reported that the use of isoflavones (particularly genistein) for the treatment of different types of cancer, including breast cancer, be promising chemopreventive agents. However, their precise mode of action in breast cancer treatment remains unclear till today. Methods Literature survey was performed for screening out the genistein with therapeutic potentials and their mode of action using published articles available at the web databases in the public domains (like PubMed, SCI Finder, Science Direct, and Google Scholar) till June 2021. Results In this review article, we have reviewed the development of novel anti-tumour strategies like inhibition of OPN at the transcriptional- or translational- level, development of monoclonal antibodies against OPN, and its downstream signalling pathways, etc. The inhibitory effect of the isoflavone(s) (e.g. genistein) on breast cancer proliferation, migration and invasion has also been reviewed. A newly elucidated anti-carcinogenic mode of action of the isoflavone including epigenetic modifications, topoisomerase inhibition and modulation of miRNA expression has also been discussed. Reports on the regulation of OPN expression by the isoflavone and the signalling pathways involved therein have been reviewed. Conclusion Taken together, the isoflavone (genistein) could be used as a promising agent for cancer chemoprevention and/or treatment, though more clinical trials are needed for its validation.
Chapter
The importance of diet and nutrition in cancer chemoprevention were indicated by a remarkable amount of evidence from experimental, clinical, and epidemiological studies. Phytoestrogens with remote structure similarities to estradiol enable to bind to receptors of estrogen. Isoflavones and lignans represent the main class of phytoestrogens, while other groups of phytoestrogens like coumestans and stilbenes are in a lower amount in food and less investigated. The most important representative of isoflavonoids are genistein, daidzein, and glycitein which accumulate to high levels in soy and legumes. Estrogen-like ability of isoflavones raise concerns in some individuals and more specifically in certain types of malignancies like breast or endometrial and prostate cancers. The available data regarding the important molecular processes of these compounds and clinical trials or observational studies were identified in chemoprevention and cancer to reveal whether there is association between soy food consumption and increase of the cancer risk. Molecular mechanisms of isoflavonoids can vary depending on their concentrations or physiological status of the cells or individuals (age, ethnicity, intestine microflora, and diet habits). Therefore, administration of these compounds has to be tailored according to the individual characteristic. Additionally, more clear recommendations can be provided by further high-quality, full powered, placebo-controlled studies considering individual characteristics as subgroups.
Article
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Article
Full-text available
Backgrounds CXC chemokine ligand 16 (CXCL16) is a soluble chemokine with a transmembrane domain, playing an important role in inflammatory regulation. NF-κB has a critical role in tumor progression. Recent studies focused on the effect of CXCL16 on tumor progression. However, few reports showed the influence of CXCL16 on lung cancer, especially in regulating NF-κB activity. Here we investigated CXCL16 expression and its clinical significance in lung cancer, as well as the effect on lung cancer cell biological characteristics by regulating NF-κB. Material/Methods CXCL16 expression in lung cancer was detected and its associations with clinical characteristics were analyzed. Proliferation and invasion of A549 and PC-9 cells was measured before and after silencing CXCL16 or inhibiting the NF-κB pathway, separately. Result The positive rate of CXCL16 in lung cancer tissue was significantly higher than that in adjacent tissue, and that in patients with lymphatic metastasis was significantly higher than that in patients without (all, P<0.05). The positive rate of CXCL16 was significantly (P<0.05) positively corrected with poor prognosis of lung cancer. Silencing CXCL16 not only suppressed proliferation and invasion of A549 and PC-9 cells, but also significantly (P<0.05) inhibited c-Rel, p105, and Rel-B in the NF-κB pathway. Inhibiting NF-κB also suppressed proliferation and invasion of A549 and PC-9 cells, which was similar to the results after silencing CXCL16. Conclusions Enhanced CXCL16 expression in lung cancer tissue promoted the proliferation and invasion of lung cancer cells. CXCL16 might promote proliferation and invasion of lung cancer by regulating the NF-κB pathway.
Article
Full-text available
Despite substantial research, the understanding of the chemopreventive mechanisms of soy isoflavones remains challenging. Promising tools, such as metabolomics, can provide now a deeper insight into their biochemical mechanisms. The purpose of this study was to offer a comprehensive assessment of the metabolic alterations induced by genistein, daidzein and a soy seed extract on estrogen responsive (MCF-7) and estrogen non-responsive breast cancer cells (MDA-MB-231), using a global metabolomic approach. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed that all test compounds induced a biphasic effect on MCF-7 cells and only a dose-dependent inhibitory effect on MDA-MB-231 cells. Proton nuclear magnetic resonance (¹H-NMR) profiling of extracellular metabolites and gas chromatography-mass spectrometry (GC-MS) profiling of intracellular metabolites confirmed that all test compounds shared similar metabolic mechanisms. Exposing MCF-7 cells to stimulatory concentrations of isoflavones led to increased intracellular levels of 6-phosphogluconate and ribose 5-phosphate, suggesting a possible upregulation of the pentose phosphate pathway. After exposure to inhibitory doses of isoflavones, a significant decrease in glucose uptake was observed, especially for MCF-7 cells. In MDA-MB-231 cells, the glutamine uptake was significantly restricted, leading to alterations in protein biosynthesis. Understanding the metabolomic alterations of isoflavones represents a step forward in considering soy and soy derivates as functional foods in breast cancer chemoprevention.
Article
Full-text available
The potential benefit of soy isoflavones in breast cancer chemoprevention, as suggested by epidemiological studies, has aroused the interest of numerous scientists for over twenty years. Although intensive work has been done in this field, the preclinical results continue to be controversial and the molecular mechanisms are far from being fully understood. The antiproliferative effect of soy isoflavones has been commonly linked to the estrogen receptor interaction, but there is growing evidence that other pathways are influenced as well. Among these, the regulation of apoptosis, cell proliferation and survival, inhibition of angiogenesis and metastasis or antioxidant properties have been recently explored using various isoflavone doses and various breast cancer cells. In this review, we offer a comprehensive perspective on the molecular mechanisms of isoflavones observed in in vitro studies, emphasizing each time the dose-effect relationship and estrogen receptor status of the cells. Furthermore, we present future research directions in this field which could provide a better understanding of the inner molecular mechanisms of soy isoflavones in breast cancer.
Article
Full-text available
Background and aims. Soy supplements are often recommended in the management of menopause symptoms. The declared content of soy supplements is commonly expressed as total isoflavones per dosage form. Given that soy isoflavones have different estrogenic potencies, pharmacokinetics and metabolism, the aim of this study was to evaluate the total isoflavone content and the aglycone profile of seven soy supplements and one soy seed extract. Label accuracy was assessed, in relation to the precise content and the recommended posology for estimating whether the optimal dose is achieved for alleviating menopause symptoms. Materials and Methods. A high performance liquid chromatography method was developed for evaluating the aglycone content (genistein, daidzein, glycitein). After extraction and acidic hydrolysis, the aglycones were separated on a C18 column, using 0.1% acetic acid and acetonitrile as mobile phases. The flow rate was 1.5mL min-1 and the UV detector wavelength was set at 260nm. A linear relationship was found in the range 5-80µg mL-1. The method was validated using the accuracy profile methodology. Results and Discussions. The total isoflavone content ranged from 6.07 to 41.68mg dosage form-1. Various aglycone profiles were obtained for each supplement which can result in a different estrogenic activity, bioavailability and finally, in a different efficiency in alleviating menopause symptoms. In most clinical trials where soy isoflavones were evaluated, little attention was paid to determining the exact aglycone profile of the employed soy extracts. Conclusions. As clinical outcomes continue to be controversial, this study highlights the need of standardization in genistein, rather than total isoflavones and labeling accuracy for soy supplements.
Article
Full-text available
Epidemiological studies have revealed that high consumption of soy products is associated with low incidences of hormone-dependent cancers, including breast and prostate cancer. Soybeans contain large amounts of isoflavones, such as the genistein and daidzain. Previously, it has been demonstrated that genistein, one of the predominant soy isoflavones, can inhibit several steps involved in carcinogenesis. It is suggested that genistein possesses pleiotropic molecular mechanisms of action including inhibition of tyrosine kinases, DNA topoisomerase II, 5α-reductase, galectin-induced G2/M arrest, protein histidine kinase, and cyclin-dependent kinases, modulation of different signaling pathways associated with the growth of cancer cells (e.g., NF-κB, Akt, MAPK), etc. Moreover, genistein is also a potent inhibitor of angiogenesis. Uncontrolled angiogenesis is considered as a key step in cancer growth, invasion, and metastasis. Genistein was found to inhibit angiogenesis through regulation of multiple pathways, such as regulation of VEGF, MMPs, EGFR expressions and NF-κB, PI3-K/Akt, ERK1/2 signaling pathways, thereby causing strong antiangiogenic effects. This review focuses on the antiangiogenic properties of soy isoflavonoids and examines their possible underlying mechanisms.
Article
Full-text available
Our previous studies demonstrate that CXCL6/CXCR6 chemokine axis induces prostate cancer progression by the AKT/mTOR signaling pathway; however, its role and mechanisms underlying invasiveness and metastasis of breast cancer are yet to be elucidated. In this investigation, CXCR6 protein expression was examined using high-density tissue microarrays and immunohistochemistry. Expression of CXCR6 shows a higher epithelial staining in breast cancer nest site and metastatic lymph node than the normal breast tissue, suggesting that CXCR6 may be involved in breast cancer (BC) development. In vitro and in vivo experiments indicate that overexpression of CXCR6 in BC cells has a marked effect on increasing cell migration, invasion and metastasis. In contrast, reduction of CXCR6 expression by shRNAs in these cells greatly reduce its invasion and metastasis ability. Mechanistic analyses show that CXCL16/CXCR6 chemokine axis is capable of modulating activation of RhoA through activating ERK1/2 signaling pathway, which then inhibits the activity of cofilin, thereby enhancing the stability of F-actin, responsible for invasiveness and metastasis of BC. Taken together, our data shows for the first time that the CXCR6 / ERK1/2/ RhoA / cofilin /F-actin pathway plays a central role in the development of BC. Targeting the signaling pathway may prove beneficial to prevent metastasis and provide a more effective therapeutic strategy for BC.
Article
Full-text available
Background: Increasing evidence argues that soluble CXCL16 promotes proliferation, migration, and invasion of cancer cells in vitro. However, the role of transmembrane or cellular CXCL16 in cancer remains relatively unknown. In this study, we determine the function of cellular CXCL16 as tumor suppressor in breast cancer cells. Methods: Expression of cellular CXCL16 in breast cancer cell lines was determined at both RNA and protein levels. In vitro and in vivo studies that overexpressed or downregulated CXCL16 were conducted in breast cancer cells. Results: We report differential expression of cellular CXCL16 in breast cancer cell lines that was negatively correlated with cell invasiveness and migration. Overexpression of CXCL16 in MDA-MB-231 cells led to a decrease in cell invasion and migration and induced apoptosis of the cells; downregulation of CXCL16 in MCF-7 cells increased cell migration and invasiveness. Consistent with the in vitro data, CXCL16 overexpression inhibited tumorigenesis in vivo. Conclusions: Cellular CXCL16 suppresses invasion and metastasis of breast cancer cells in vitro and inhibits tumorigenesis in vivo. Targeting of cellular CXCL16 expression is a potential therapeutic strategy for breast cancer.
Article
Estrogen receptors (ERs) α and β are distributed in most tissues of women and men. ERs are bound by estradiol (E2), a natural hormone, and mediate the pleiotropic and tissue-specific effects of E2, such as proliferation of breast epithelial cells or protection and differentiation of neuronal cells. Numerous environmental molecules, called endocrine disrupting compounds, also interact with ERs. Phytoestrogens belong to this large family and are considered potent therapeutic molecules that act through their selective estrogen receptor modulator (SERM) activity. Using breast cancer cell lines as a model of estrogen-dependent proliferation and a stably ER-expressing PC12 cell line as a model of neuronal differentiating cells, we studied the SERM activity of major dietary compounds, such as apigenin, liquiritigenin, daidzein, genistein, coumestrol, resveratrol and zearalenone. The ability of these compounds to induce ER-transactivation and breast cancer cell proliferation and enhance Nerve Growth Factor (NGF) -induced neuritogenesis was assessed. Surprisingly, although all compounds were able to activate the ER through an estrogen responsive element reporter gene, they showed differential activity toward proliferation or differentiation. Apigenin and resveratrol showed a partial or no proliferative effect on breast cancer cells but fully contributed to the neuritogenesis effect of NGF. However, daidzein and zearalenone showed full effects on cellular proliferation but did not induce cellular differentiation. In summary, our results suggest that the therapeutic potential of phytoestrogens can diverge depending on the molecule and the phenotype considered. Hence, apigenin and resveratrol might be used in the development of therapeutics for breast cancer and brain diseases.
Article
The chemokine CXCL16 and its receptor, C-X-C chemokine receptor (CXCR6), affect tumor progression through different pathways, including leukocyte recruitment and function, cellular senescence, tumor cell proliferation, survival, invasion, and metastasis. We examined how the expression of CXCL16/CXCR6 in prostate cancer (PC) was related to clinicopathological features and activation of inflammatory cells. Tissue microarrays from 535 patients were constructed from tumor epithelial and tumor stromal areas of primary PC. Immunohistochemistry was used to evaluate the expression of CXCL16/CXCR6, CD3(+) T cells (CD4(+), CD8(+)), and CD20(+) B cells. Survival analyses were used to evaluate their prognostic impact. Expression of CXCL16 in PC cell lines (DU145 and PC3) and the effect on proliferation and migration were examined. High expression levels of CXCL16 [hazard ratio (HR), 2.52; 95% CI, 1.12-5.68; P = 0.026] and CXCR6 (HR, 2.29; 95% CI, 1.10-4.82; P = 0.028) were each independent predictors for clinical failure. High co-expression of CXCL16 and CXCR6 (HR, 5.1; 95% CI, 1-15.9; P = 0.05) was associated with negative prognostic factors, such as Gleason grade 4 + 3, Gleason score ≥7, vascular infiltration, and positive surgical margins. As a conclusion, high protein expression of CXCL16 and high protein co-expression of CXCL16/CXCR6 in PC were independent predictors for a worse clinical outcome. Copyright © 2015 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.