ArticlePDF Available

Curcumin inhibits TGF-??1-induced MMP-9 and invasion through ERK and Smad signaling in breast cancer MDA- MB-231 cells

Authors:

Abstract

Objective: To evaluate the effects of curcumin on matrixmetalloproteinase-9 (MMP-9) and invasion ability induced by transforming growth factor-β1 (TGF-β1) in MDA-MB-231 cells and potential mechanisms. Methods: Human breast cancer MDA- MB-231 cells were used with the CCK-8 assay to measure the cytotoxicity of curcumin. After treatment with 10 ng/ml TGF-β1, with or without curcumin (≤10 μM), cell invasion was checked by transwell chamber. The effects of curcumin on TGF-β1-stimulated MMP-9 and phosphorylation of Smad2, extracellular-regulated kinase (ERK), and p38 mitogen activated protein kinases (p38MAPK) were examined by Western blotting. Supernatant liquid were collected to analyze the activity of MMP-9 via zymography. Following treatment with PD98059, a specific inhibitor of ERK, and SB203580, a specific inhibitor of p38MAPK, Western blotting and zymography were employed to examine MMP-9 expression and activity, respectively. Results: Low dose curcumin (≤10 μM) did not show any obvious toxicity to the cells, while 0~10 μmol/L caused a concentration-dependent reduction in cell invasion provoked by TGF-β1. Curcumin also markedly inhibited TGF-β1-regulated MMP-9 and activation of Smad2, ERK1/2 and p38 in a dose- and time-dependent manner. Additionally, PD98059, but not SB203580, showed a similar pattern of inhibition of MMP-9 expression. Conclusion: Curcumin inhibited TGF-β1-stimulated MMP-9 and the invasive phenotype in MDA-MB-231 cells, possibly associated with TGF-β/Smad and TGF-β/ERK signaling.
Asian Pacic Journal of Cancer Prevention, Vol 13, 2012 5709
DOI:http://dx.doi.org/10.7314/APJCP.2012.13.11.5709
Curcumin Inhibits TGF-β1-Induced MMP -9 and Invasion MDA-MB-231 Cells through ERK, Smad Signaling
Asian Pacic J Cancer Prev, 13 (11), 5709-5714
Introduction
Breast cancer is one of most common malignant tumor
in global female. And the leading cause of morbidity and
mortality in breast cancer patients is cancer metastases, but
not the primary tumor (Porter, 2009; Sanchez-Zamorano
et al., 2011). It has been demonstrated that tumor cell
invasion is the key event in the metastatic steps (Geiger
et al., 2009). However, recent studies show that cancer
progression toward metastasis is not entirely dependent on
tumor cells themselves, but the result of the interactions
of tumor cells and the primary tumor microenvironment
(Allen et al., 2011). Cancer cells, specially those at the
edges of the tumor, can attract and stimulate host cells
to secret many kinds of factors (TGF-β1, IL-1, TNF-α,
etc.) in the tumor microenvironment. Those factors can
generate a series of invasion-related biological effects
through the tumor cell signal transduction pathways, and
then cause the cells to separate from the primary lesion
focus and result in local invasion and distant mestastasis
nally (Allen et al., 2011). So far, novel ndings indicates
that TGF-β1 is one of the factors which could be highly
1Department of Pathology, Chongqing Medical University, Chongqing 2Department of Anesthesiology, Peking University Third
Hospital (PUTH), Beijing, China *For correspondence: cydcyj@163.com
Abstract
Objective: To evaluate the effects of curcumin on matrixmetalloproteinase-9 (MMP-9) and invasion ability
induced by transforming growth factor-β1 (TGF-β1) in MDA-MB-231 cells and potential mechanisms. Methods:
Human breast cancer MDA- MB-231 cells were used with the CCK-8 assay to measure the cytotoxicity of
curcumin. After treatment with 10 ng/ml TGF-β1, with or without curcumin (≤10 μM), cell invasion was checked
by transwell chamber. The effects of curcumin on TGF-β1-stimulated MMP-9 and phosphorylation of Smad2,
extracellular-regulated kinase (ERK), and p38 mitogen activated protein kinases (p38MAPK) were examined by
Western blotting. Supernatant liquid were collected to analyze the activity of MMP-9 via zymography. Following
treatment with PD98059, a specic inhibitor of ERK, and SB203580, a specic inhibitor of p38MAPK, Western
blotting and zymography were employed to examine MMP-9 expression and activity, respectively. Results: Low
dose curcumin (≤10 μM) did not show any obvious toxicity to the cells, while 0~10 μmol/L caused a concentration–
dependent reduction in cell invasion provoked by TGF-β1. Curcumin also markedly inhibited TGF-β1-regulated
MMP-9 and activation of Smad2, ERK1/2 and p38 in a dose- and time-dependent manner. Additionally, PD98059,
but not SB203580, showed a similar pattern of inhibition of MMP-9 expression. Conclusion: Curcumin inhibited
TGF-β1-stimulated MMP-9 and the invasive phenotype in MDA-MB-231 cells, possibly associated with TGF-β/
Smad and TGF-β /ERK signaling.
Keywords: Curcumin - breast cancer - TGF-β1 - MMP-9 - MAPKs - invasion
RESEARCH ARTICLE
Curcumin Inhibits TGF-β1-Induced MMP-9 and Invasion
through ERK and Smad Signaling in Breast Cancer MDA-
MB-231 Cells
Na Mo1, Zheng-Qian Li2, Jing Li1, You-De Cao1*
relevant to breast cancer invasion and metastasis (Perera
et al., 2010).
TGF-β1 is known to be one of the TGF-β superfamily
members that plays a dual role in breast cancer generation
and progression. In the early stages, TGF-β1 acts as
a tumor suppressor, nevertheless, it promotes cancer
invasion and metastasis in later stages (Biswas et al.,
2007). After binding to TGF-β1, the type II TGF-β1
receptor recruits and phosphorylates the type I receptor.
The activated type I receptor subsequently initiates the
activation of Receptor-associated Smads (R-Smads), then
the latter forms heteromeric complexes with common
mediator Smad4 (co-Smad4) and translocate into the
nucleus. The Smad complexes regulate specic target
genes by directly or indirectly combining and interacting
with transcription factors, co-repressors, and co-activators
(Smith et al., 2012). In addition to the classical TGF-β/
Smad signaling pathway, TGF-β1 can also directly
activate non-Smad signaling pathways (Zhang, 2009).
Previous reports have shown the direct function of MAPK
pathways, including c-Jun N-terminal kinase (JNK),
extracellular- regulated kinase ERK, and p38 MAPK, in
Na Mo et al
Asian Pacic Journal of Cancer Prevention, Vol 13, 2012
5710
signal transduction of TGF-β1-regulated cell migration
and invasion (Sana et al., 2007).
Researchers have demonstrated that the serum TGF-β1
level in early-stage breast cancer patients is uplifted
and the high level of TGF-β1 has positive correlation
with the effects of anti-tumor. However, it accelerates
cancer invasion and formation of metastases in late-stage
breast cancer (Cheung, 2007). Serra R et al. have found
that radiation and chemotherapy can quicken tumor
cells diffusion in mouse models of breast cancer. The
experimental datas indicate that the levels of TGF-β1
in those mouse models have been rised remarkably
. In contrast, there is no far metastasis in the mouse
models with low level of TGF-β1 (Serra et al., 2005).
Therefore, antagonism of TGF-β1 signaling may provide
a therapeutic target for late-stage breast cancer, blocking
metastasis without detrimental side effects.
Curcumin is a natural phenolic pigment extracted from
the roots of turmeric. Numerous studies have reported that
it has positive pharmacological effects, such as anti-tumor,
anti-oxidation, anti-inammation, anti-rheumatism and so
on (Yan et al., 2012). The U.S. National Cancer Institute
has listed it as the third-generation anti-cancer drug to
study, so that curcumin will likely to be a promising
clinical anti-cancer drug (Park et al., 2008). Nevertheless,
the precise molecular mechanisms underlying its anti-
tumor invasion and metastasis are not entirely clear (Park
et al., 2008). Most of the reports pay more attention to the
anti-brosis activity of curcumin on TGF-β1-stimulated
organs (Tubulointerstitial, corneal, liver, etc.) brosis via
blocking TGF-β signing pathway (Smith et al., 2010; Yao
et al., 2012; Zhang et al., 2012). Kim et al. have conrmed
that curcumin inhibites TGF-β1-induced MMPs in mouse
keratinocytes (Santibáñez et al., 2000; Santibáñez et al.,
2002), but the effects on breast cancer MDA-MB -231
have not been reported.
Therefore, the present study investigated the effect
of curcumin on the exogenous TGF-β1–stimulated
expression and activity of MMP-9 in human breast
cancer MDA-MB-231 cells. Furthermore, the underlying
mechanisms were also probed.
Materials and Methods
Cell culture
MDA-MB-231 cells were incubated with RPMI-1640
medium (Gibco, California, USA) containing 10% fetal
calf serum at 37°C in 5% CO2 incubator. Cells were
cultured in serum-free media for 24h to synchronize
cell growth before the experiments, then the media
were exchanged for fresh serum-free medium, treated
with various agents at the concentrations specied, and
cells were harvested at different time points for various
analyses.
Cytotoxicity assay
MDA-MB-231 cells were trypsinized and seeded in
96-well plates at 5 × 103 cells/well. After 24 h, escalating
doses of curcumin (Sigma, USA) were added, and
incubated for another 24 h, 48h, and 72h respectively.
Cells without any treatment were used as control. Then,
10 μL CCK-8 (Dojindo, Tokyo, Japan) solution in culture
medium was added to each well. Plates were incubated
for an additional 2 h. The optical density of each well was
measured using microplate absorbance reader at a 450
nm wavelength. Cell viability was calculated as follows.
The cell survival rate (%) = [(A Treatment group- A blank
wells) / (A negative control group-A blank wells)] × 100%.
Invasion assay
The MDA-MB-231 cells invasion behavior with
or without indicated treatment was tested by Matrigel
transwell system as described previously (Ye et al., 2012) .
After cultured in 6 well plates for 48h, the cells in different
experimental groups were trypsinized, centrifuged, and
resuspended at 1×106 cells/mL in serum-free medium
respectively. 100μl cell suspension per well were seeded
onto the upper wells of transwells (8-µm-diameter pores;
Millipore), which precoated with Matrigel (0.5 mg/Ml,
BD Biosciences Discovery Labware). Lower chamber of
the transwells contained the medium containing 10% FBS
as chemoattractant. After 8 h of incubation, the cells on
the upper chamber were carefully wiped with the cotton
swab. The wells were washed 3 times with PBS, then
xed with 4% paraformaldehyde, and stained with crystal
violet solution (Sigma Chemical, USA). The cells on the
lower surface of the membrane were counted under a light
microscope (magnication, ×100). The experiments were
performed three times, each time in triplicate.
Gelatin zymography
Cells in the logarithmic phase were seeded in 6-well
plate at the desity of 3 × 105 cells per well. After incubated
in serum-free medium with or without curcumin (2.5, 7.5
and 10 μM) and 10 μM TGF-β1 treatment for 48h. The
supernatants were collected, and gelatin zymography
assay was performed as described formerly (Zayani et al.
2012). After electrophoresis, the gels were washed three
times with renaturing buffer containing 50 mM Tris–HCl,
5 mM CaCl2 and 2.5% Triton X-100 (v/v), pH 7.5 for
30 min, followed by a brief rinsing with washing buffer
(50 mM Tris–HCl , 5 mM CaCl2), pH 7.5. Then the gels
were incubated at 37°C for 42 h in developing buffer
containing 50 mM Tris–HCl, 5 mM CaCl2, 0.2 M NaCl,
and 0.02% Brij 35, pH 7.5. The gels were subsequently
stained with 0.25% Coomassie Brilliant Blue (G250)
followed by destaining with a solution containing 10%
acetic acid and 20% methanol. Enzyme-digested regions
were visualized as light bands against a dark background.
Zones of enzymatic activity were regarded as negatively
stained bands.
Western blotting
Cells in experimental groups were collected and
lysed in RIPA buffer respectively. Supernatants of the
cell lysates were used in the western blot analysis for
MMP-9 (Bioss, Beijing, China), β-actin (Beyotime,
Beijing, China), and phosphorylation levels of ERK1/2,
Smad2, p38MAPK (Cell signing technology, USA). After
electrophoresis and trarsmembrane, the PVDF membranes
(Millipore, USA)containing the proteins were blocked
with 5% bovine serum albumin in TBST buffer (0.01%
Asian Pacic Journal of Cancer Prevention, Vol 13, 2012 5711
DOI:http://dx.doi.org/10.7314/APJCP.2012.13.11.5709
Curcumin Inhibits TGF-β1-Induced MMP -9 and Invasion MDA-MB-231 Cells through ERK, Smad Signaling
Figure 1. The Cell Toxicity of MDA-MB-231 Cells
Treated by Curcumin. * P<0.05 vs. 0 μM Cur (24h), P<0.05
vs. 0 μM Cur (48h), P<0.05 vs. 0 μM Cur (72h)
Figure 2. Effect of Curcumin on TGF-β1-induced
Invasion Ability of MDA-MB-231 Cells in vitro (crystal
violet ×100) (a) MDA-MB-231 cells were incubated with
different doses of Cur ,with or without TGF-β (10 ng/ml)
for 48h, then checked invasion ability by Transwell assay. (b)
Quantitation of the cells which invasive matrigel to lower surface
of the membrane by cell counting under microscope (×100)
(mean±SEM from 3 independent tests).*P<0.05 vs. control,
**P<0.05 vs. TGF-β1 alone
Figure 3. Effect of Curcumin on MMP-9 Protein
Expression Activated by TGF-β1 (a, b) Cell were incubated
with different doses of curcumin and with or without TGF-β1
for 48h , and the MMP-9 Protein expression was checked by
Western blotting . (mean ± SEM from 3 separate tests) *P<0.05
vs. control, **P<0.05 vs. TGF-β1 alone. (c, d) MDA-MB-231
cells were treated with 10μM Cur, followed by TGF-β1 for
different times, and the MMP-9 Protein expression was analyzed
by Western blotting (mean ± SEM from 3 separate tests) *P<0.05
vs. control, **P<0.05 vs. TGF-β1 alone
a b
c d
Tween 20 in TBS) at room temperature(RT) for about 2h,
following by incubated with the primary antibody (i.e.,
specic antibodies against the target proteins described
above) in TBST at 4℃ overnight. The membranes were
washed three times in TBST and subsequently covered
with anti-rabbit HRP-conjugated goat anti-rabbit IgG
(Bioss, Beijing, China) or goat anti-mouse IgG (Beyotime,
Beijing, China) at RT for 2h in TBST buffer. After a
2h incubation, all blots were washed three times and
ECL reagents (Beyotime, Beijing, China) were used for
development.
Statistical analysis
Data are presented as means ± SEM and analyzed for
statistical signicance using independent samples T-test.
All Statistical analysis was performed with SPSS for
Windows (version 14.0; SPSS, Chicago, IL). Statistical
signicance was considered as P <0.05.
Results
The cytotoxicity effect of curcumin on the growth of MDA-
MB-231 cells
To evaluate cytotoxicity of curcumin on the growth
of breast cancer cells, MDA- MB-231 cells were treated
with 5, 10, 15, 20, 30 and 50 μM curcumin for 24h, 48h,
72h respectively and then cell viability was detected by
CCK-8 assay. As shown in Figure1, low-dose curcumin
(≤10 μM) did not affect the viability of MDA-MB-231
cells, and survival rates of all the low-dose groups had
exceeded 90% (except 10 μM curcumin for 72 h). But
when the concentrations was above 10μM, curcumin
time- and dose- dependently inhibited the growth of
MDA-MB-231 cells. Therefore, the cells were treated
with selected doses (≤ 10μM) for no more than 48 hours
in subsequent experiments.
Effects of curcumin on TGF-β1-induced invasiveness of
MDA -MB -231 cells
We next examined the effect of curcumin on TGF-β1-
induced cell invasion in MDA-MB-231 breast cancer cells
using the transwell chamber assay. Our results showed that
the invasiveness of MDA-MB-231 cells was increased by
TGF-β1 treatment (Figure 2). On the other hand, TGF-
β1-stimulated invasiveness of cells was decreased by
curcumin in a dose-dependent manner (Figure 2). These
results were inconsistent with the results of the wound
healing assay (data did not shown). This suggests that
curcumin can prevent the TGF-β1-induced invasion in
MDA-MB-231 cells.
Effects of curcumin on TGF-β1-Mediated MMP-9 protein
expression and activity in MDA -MB -231 cells
We examined whether curcumin involved with the
TGF-β1-induced MMP-9 protein expression. After
pretreatment with different concentrations of curcumin
for 30 min, the cells were cultured with TGF-β1
and various doses of curcumin for 48h. Western blot
analyses revealed that TGF-β1-induced MMP-9 protein
expression was signicantly decreased by curcumin in
a dose-dependent manner. The level of MMP-9 protein
expressions was increased to 1.93-fold of the control
level by 10 nM TGF-β1 treatment, while the TGF-β1-
induced MMP-9 protein expressions was decreased to
81.5%, 70.4%, 55.0% of the 10 nM TGF-β1 group level
by 5, 7.5 and 10μM curcumin treatment, respectively
(Figure 3A, 3B). So, 10 μM curcumin has the maximal
inhibitory effect. After pretreatment with 10μM curcumin
Na Mo et al
Asian Pacic Journal of Cancer Prevention, Vol 13, 2012
5712
for 30 min, the cells were treated with 10 nM TGF-β1
and 10μM curcumin for 12h, 24h, 48h, respectively. Our
results showed that the TGF-β1-induced MMP-9 protein
expressions were evidently suppressed by curcumin in
a time-dependent way. The TGF-β1-induced MMP-9
protein expressions was decreased by 77.5%, 60.4%,
58.4% of the 10 nM TGF-β1 group level in 12h, 24h,
48h respectively. Therefore, curcumin had the best
inhibitory effect at 48h. As exhibited in Figure 3 C and
D, treatment with 10 nM TGF-β1 for 24h led to increase
enzymatic activity of MMP-9. In contrast, curcumin dose-
dependently inhibited this effect, with 10 μM curcumin
showing optimum inhibitory effect.
Effect of TGF-β1 on phosphorlation of Smad2, ERK and
p38MAPK in breast cancer MDA-MB-231 cells
As shown in Figure 4, 10μM TGF-β1 stimulated
phosphorylation of Smad2, ERK and p38MAPK as early
as 15 minutes while p-Smad2 peaked at 30 minutes. And
the p-ERK and p-p38MAPK reached maximum at 60 min.
The levels of total Smad2, ERK and p38 did not altered.
Effect of curcumin on activation of Smad and MAPK
Pathway by TGF-β1
Cells were treated with 10μM curcumin for 0, 6,
12, and 24 h, with or without inducing of TGF-β1 for
an additional 15 min. Western blot analyses indicated
that TGF-β1 markedly increased activation of Smad2,
ERK1/2, and p38MAPK at 12h and 24h, but did not
increase levels of total Smad2, ERK1/2, p38MAPK.
Curcumin at 10μM exhibited signicantly inhibitive effect
on TGF-β1-induced phosphorylation of Smad2, ERK1/2,
p38MAPK at different intervals, and the 12 h treated group
was more effective. Compared with the same time point
of TGF-β1 alone treatment group, the difference was
signicant (P <0.001) as shown in Figure 5 A and B.
Next, cells were incubated with different concentrations
(0, 5, 7.5, 10 μM) of curcumin for 12 h, followed by
treatment of TGF-β1 for an additional 15 min. The results
revealed that TGF-β1 at 10μM dependently induced
p-Smad2, p-ERK1/2 and p-p38MAPK when compared
with constant levels of total Smad2, ERK1/2, p38 MAPK.
Curcumin significantly inhibited TGF-β1-induced
phosphorlation of Smad2, ERK1/2 and P38MAPK in
MDA-MB-231 cells at protein levels,and the inhibition
effect was characterized by a concentration-dependent
way, and the dose of 10 μM was regarded as the best
inhibition effect (Figure 5 C, D)
Effect of PD98059, SB202580, curcumin on MMP-9
enzymatic activity and expression by TGF-β1
Cells were treated with curcumin (10μM) , PD98059
(30μM) , SB202580 (20μM) for 48 h, combination with
or without treatment of TGF-β1 (10 ng/ml). The inhibitory
effect of PD98059 on MMP-9 was similar to curcumin,
whereas SB202580 group shown no signicant change
when compared with the TGF-β1 alone group (Figure 6).
Discussion
About one-third of women with breast cancer develop
distant metastasis and ultimately die worldwide each year
(Allen et al., 2011; Nasser et al., 2012). Thus, metastatic
breast cancer has been thought to be the principal
challenge for the effective treatment and prevention
Figure 4. Effects of TGF-β1 on p-smad2, p-ERK1/2,
p-p38 Expression in Different Time Periods. MDA-
MB-231 cells were treated with TGF-β1 (10 ng/ml) for the
indicated times, and the phosphorylation of Smad2< ERK1/2 and
p38 was analyzed by Western blotting. The same membrane was
retested with anti-Smad2<ERK1/2 and p38 antibody
Figure 5. Effects of Curcumin on p-smad2, p-ERK1/2,
p-p38 Expression Activated by TGF-β1. (a) (b) MDA-
MB-231 cells were treated with 10μM Cur for different times,
with or without TGF-β1 for an additional 15min, and the
phosphorylation of Smad2, ERK1/2, p-p38 was analyzed by
Western blotting (mean ± SEM from 3 separate tests) *P<0.05 vs.
TGF-β1 alone. (c) (d) Cells were incubated with different doses
of Cur for 12h,and with or without TGF-β1 for an additional
15min , and the phosphorylation of Smad2, ERK1/2, p-p38 was
checked by Western blotting . (mean ± SEM from 3 independent
tests) *P<0.05 vs. control, **P<0.05 vs. TGF-β1 alone
a b
c d
Figure 6. Effects of PD98059, SB202580, and Curcumin
on TGF-β1-induced MMP-9 in MDA-MB-231 Cells.
The MMP-9 expression and enzymatic activity were analyzed by
Western blot and zymography, respectively (*P<0.05 vs. control
P<0.05 vs. TGF-β1 alone)
Asian Pacic Journal of Cancer Prevention, Vol 13, 2012 5713
DOI:http://dx.doi.org/10.7314/APJCP.2012.13.11.5709
Curcumin Inhibits TGF-β1-Induced MMP -9 and Invasion MDA-MB-231 Cells through ERK, Smad Signaling
(Hortobagyi et al., 2002). Extensive evidence now
indicates that degradation of extracelluar matrix assisting
cancer cell to invade neighbouring tissue, blood vessels
and spread to other sites is the essential process of distant
metastases in invasive breast cancer (Hassan et al., 2012).
Reportedly, MMP-9 plays a signicant role in breast
cancer invasion and metastasis via degrading type IV
collagen-rich extracelluar matrix (Chen et al., 2011) , and
also be regulated by TGF-β1, epidermal growth factor
(EGF), broblast growth factor (FGF), Nerve growth
factor (NGF), vascular endothelial growth factor (VEGF),
etc., which secreted by cancer cells and/or host cells in
the tumor microenvironment (Chou et al., 2006; Belotti
et al., 2008; Blair et al., 2011). Among those factors,
TGF-β1 generated by autocrine and paracrine is high
related to malignant tumor (Na et al., 2010). And TGF-β
signing pathway is of great importance of breast cancer
invasivness and metastasis (Imamura et al., 2012). Based
on these reports, compounds that can suppress TGF-β1-
induced MMP-9 are applied for the treatment of metastatic
breast cancer. Many researchers focus mostly on the anti-
brotic effects of curcumin via inhibiting TGF-β signing
pathway (Smith et al., 2010; Yao et al., 2012; Zhang et al.,
2012). However, whether curcumin can suppress TGF-β1-
stimulated MMP-9 expression was not clear.
In accordance with previous reports (Yodkeeree et al.,
2010), invasion assay in our study shown that curcumin at
a nontoxic concentration makedly decreased invasiveness
in response to TGF-β1 in a dose-dependent manner. Our
results demonstrated that the levels of MMP-9 protein
expression and enzymatic activity were signicantly
increased stimulated by TGF-β1 in the MDA-MB-231
breast cancer cells, and this increase can be inhibited
obviously by curcumin. So, we came to the conclusion
that TGF-β1-stimulated MMP-9 expression and activity
might mediate cell invasion during wound healing and
curcumin inhibited this process in MDA-MB-231 cells.
In classical TGF-β/Smad pathway, TGF-β1 signal
through TGF-β/receptor serine- threonine kinases. The
activated receptor complex phosphorylated receptor-
regulated Smads (R-Smads), the latter forms heteromeric
complexes with Smad4 that translocate into the nucleus
and regulate transcription of specic target genes (Zhang,
2009). Besides TGF-β/Smad pathway, TGF-β1 can also
directly activate non-Smad signaling pathways, including
the MAPKs (JNK, ERK, p38) (Smith et al., 2012). In
other words, different transcriptional responses to TGF-β1
depend on activation of either or both Smad and Smad-
independent pathways. So far, several study revealed
that TGF-β1 might regulate MMP-9 expression through
rapidly activating intracellular Smads and MAPKs signing
pathway (Chou et al., 2006). However, the previous
study have shown that TGF-β/MAPKs pathway which
participate in regulating expression and activity of
MMP-9 has specicity in defferent types of cells (Kim et
al., 2005; Dziembowska et al., 2007; Szuster-Ciesielska
et al., 2011). For example, Hsieh et al. have found that
TGF-β1 induced MMP-9 expression is associated with
the ERK signing pathway in gastric cancer cells (Hsieh
et al., 2010). Nevertheless, this process is inconclusive
in breast cancer MDA-MB-231 cells. Some researchers
have also used kinase inhibitors to implicate both p38 and
ERK signing pathways in TGF-β1-mediated regulation
of MMP-9 (Iiunga et al., 2004). However, other results
have revealed that p38 inhibitor in minimal effective
dose has no effects on TGF-β1-induced MMP-9, while
excess p38 inhibitors do. The subsequent research have
found that excess p38 inhibitors not only decreased
phosphorylation of p38, but also can effectively block the
activation of TGF-β receptors kinases (Sana et al., 2007).
Our results indicated that non-toxic dose of curcumin
(≤10μM) markedly inhibited phosphorylation of Smad2,
ERK1/2, p38 mediated by TGF-β1 in a dose- and time-
dependent way. Furthermore, PD98059 and curcumin had
the similar inhibitory effects on TGF-β1-induced MMP-9.
Nevertheless, the lowest effect concentration of SB203580
did not affect regulation of MMP-9. These results were
consistent with the results have reported by Sana A
et al.. Accordingly, our ndings argued that curcumin
perhaps down-regulated TGF-β-induced MMP-9 via a
mechanism involving ERK, Smad2 but not p38MAPK
in MDA-MB-231 cells.
Moreover, previous studys have revealed that the
p38 pathway might enhance cell metastasis ability by
regulating actin remodeling factor HSP27 (Hedges et
al., 1999) and affecting actin polymerization and cell
contractility (Srinivasan et al. 2008). ERK may regulate
cell motility by preventing formation of extensive actin
stress bers via suppression of tropomyosin induction
by TGF-β1 or through inhibition of RhoA-Rho kinase
pathway (Bakin et al., 2004; Helfman et al., 2005). So,
perhaps activation of ERK, Smads and p38 MAPK signing
pathways is all required for the suppression of curcumin
on TGF-β1-mediated cell migration. This speculative
conclusion will also be our future research directions.
Our data revealed that the change of ERK1/2 and
Smad2 phosphorylation was the key point of the anti-
invasion effect of curcumin. Although our results also
found that curcumin was able to reduce TGF-β RII
expression in MDA-MB-231 cells (data does not show).
However, as the lack of specic protein phosphatase
inhibitors, we cannot exclude the possibility that
curcumin inhibits TGF-β1-stimulated MMP-9 via directly
suppressing activiation of the TGF-β receptors which acts
as an upstream regulator of ERK and Smad pathway .
In conclusion: In the present study, we explored
the inhibition action of curcumin on TGF-β1–induced
invasion in human breast cancer MDA-MB-231 cells and
disclosed the potential mechanisms of the anti–invasion
and metastasis effect. We demonstrated that (1) Curcumin
dose-dependently inhibited the invasion ablity induced by
TGF-β1 in MDA-MB-231 cells. (2) Curcumin inhibited
TGF-β1-induced MMP-9 protein expression and activity
in MDA-MB-231 cells in a time- and concentration- way.
(3) Curcumin time- and dose-dependently inhibited
Smad2, ERK1/2, p38MAPK phosphorylation induced
by TGF-β1. (4) PD98059 and curcumin had the similar
suppression effect on TGF-β1-induced MMP-9 protein
expression and activity. All together, these findings
highlight the protable effect of curcumin, and it serves
as an anti-MMP-9 factor through inhibition of the TGF-β/
Smad and TGF-β/Erk signaling pathway.
Na Mo et al
Asian Pacic Journal of Cancer Prevention, Vol 13, 2012
5714
References
Allen M, Louise Jones J (2011). Jekyll and Hyde: the role of the
microenvironment on the progression of cancer. J Pathol,
223, 162-76.
Bakin AV, Sana A, Rinehart C, et al (2004). A critical role
of tropomyosins in TGF-beta regulation of the actin
cytoskeleton andcell motility in epithelial cells. Daroqui
C, Darbary H, Helfman DM. Mol Biol Cell, 15, 4682-94.
Belotti D, Calcagno C, Garofalo A (2008). Vascular endothelial
growth factor stimulates organ-specific host matrix
metalloproteinase-9 expression and ovarian cancer invasion.
Mol Cancer Res, 6, 525-34.
Biswas S, Guix M, Rinehart C, et al (2007). Inhibition of TGF-
beta with neutralizing antibodies prevents radiation-induced
acceleration of metastatic cancer progression. J Clin Invest,
117, 1305-13.
Blair KJ, Kiang A, Wang-Rodriguez J, et al (2011). EGF and
bFGF promote invasion that is modulated by PI3/Akt
kinase and Erk in vestibular schwannoma. Otol Neurotol,
32, 308-14.
Chen P, Lu N, Ling Y, Chen Y, et al (2011). Inhibitory effects of
wogonin on the invasion of human breast carcinoma cells
by downregulating the expression and activity of matrix me
talloproteinase-9. Toxicology, 282, 122-8.
Cheung KL (2007). Endocrine therapy for breast cancer: an
overview. Breast, 16, 327-43.
Chou YT, Wang H, Chen Y (2006). Cited2 modulates TGF-beta-
mediated upregulation of MMP9. Oncogene, 25, 5547-60.
Dziembowska M, Danilkiewicz M, Wesolowska A (2007).
Cross-talk between Smad and p38 MAPK signalling in
transforming growth factor beta signal transduction in
human glioblastoma cells. Biochem Biophys Res Commun
, 354, 1101-6.
Hassan ZK, Daghestani MH (2012). Curcumin effect on MMPs
and TIMPs genes in a breast cancer cell line. Asian Pac J
Cancer Prev, 13, 3259-64.
Hortobagyi GN (2002). The status of breast cancer management:
challenges and opportunities. Breast Cancer Res Treat, 75,
S61-5, discussion S57-9.
Hsieh HL, Wang HH, Wu WB, et al (2010). Transforming
growth factor-β1 induces matrix metalloproteinase-9 and cell
migration in astrocytes: roles of ROS-dependent ERK-and
JNK-NF-κB pathways. J Neuroinammation, s, 88.
Hedges JC, Dechert MA, Yamboliev IA, et al (1999).A role
for p38(MAPK)/HSP27 pathway in smooth muscle cell
migration. J Biol Chem, 274, 24211-9.
Helfman DM, Pawlak G (2004). Myosin light chain kinase and
acto-myosin contractility modulate activation of theERK
cascade downstream of oncogenic Ras. J Cell Biochem,
95, 1069-80.
IIunga K, Nishiura R, Inada H (2004). Co-stimulation of human
breast cancer cells with transforming growth factor-beta and
tenascin-C enhances matrix metalloproteinase-9 expression
and cancer cell invasion. Int J Exp Pathol, 85, 373-9
Imamura T, Hikita A, Inoue Y (2012). The roles of TGF-β
signaling in carcinogenesis and breast cancer metastasis.
Breast Cancer, 19, 118-24.
Geiger TR, Peeper DS (2009). Metastasis mechanisms. Biochim
Biophys Acta, 1796, 293-308.
Kim HS, Luo L, Pugfelder SC, Li DQ (2005). Doxycycline
inhibits TGF-beta1-induced MMP-9 via Smad and
MAPK pathways in human corneal epithelial cells. Invest
Ophthalmol Vis Sci, 46, 840-8.
Na D, Lv ZD, Liu FN, et al (2010). Transforming growth factor
beta1 produced in autocrine/ paracrine manner affects the
morphology and function of mesothelial cells and promotes
peritoneal carcinomatosis. Int J Mol Med, 26, 325-32.
Harhra NA, Basaleem HO (2012). Trends of breast cancer and
its management in the last twenty years in aden and adjacent
governorates, yemen. Asian Pac J Cancer Prev, 13, 4347-51
Park J, Ayyappan V, Bae EK, et al (2008). Curcumin in
combination with bortezomib synergistically induced
apoptosis in human multiple myeloma U266 cells. Mol
Oncol, 2, 317-26.
Perera M, Tsang CS, Distel RJ, et al (2010). TGF-beta1
interactome: metastasis and beyond. Cancer Genomics
Proteomics, 7, 217-29.
Porter PL(2009). Global trends in breast cancer incidence and
mortality. Salud Publica Mex, 51, S141-6.
Sana A, Vandette E, Bakin AV (2007). ALK5 promotes tumor
angiogenesis by upregulating matrix metalloproteinase-9 in
tumor cells. Oncogene, 26, 2407-22.
Sanchez-Zamorano LM, Flores-Luna L, Angeles-Llerenas A et al
(2011). Healthy lifestyle on the risk of breast cancer. Cancer
Epidemiol Biomarkers Prev, 20, 912-22.
Santibáñez JF, Guerrero J, Quintanilla M, et al (2002).
Transforming growth factor-beta1 modulates matrix
metalloproteinase-9 production through the Ras/MAPK
signaling pathway in transformed keratinocytes. Biochem
Biophys Res Commun, 16, 267-73.
Santibáñez JF, Quintanilla M, Martínez J (2000). Genistein
and curcumin block TGF-beta 1-induced u-PA expression
and migratory and invasive phenotype in mouse epidermal
keratinocytes. Nutr Cancer, 37, 49-54.
Serra R, Crowley MR (2005). Mouse models of transforming
growth factor beta impact in breast development and cancer.
Endocr Relat Cancer, 12, 749-60.
Smith AL, Robin TP, Ford HL (2012). Molecular Pathways:
Targeting the TGF-beta Pathway for Cancer Therapy. Clin
Cancer Res, 18, 4514-21.
Smith MR, Gangireddy SR, Narala VR, et al (2010). Curcumin
inhibits brosis-related effects in IPF broblasts and in mice
following bleomycin-induced lung injury. Am J Physiol Lung
Cell Mol Physiol, 298, L616-25.
Srinivasan R, Forman S, Quinlan RA, et al (2008).Regulation
of contractility by Hsp27 and Hic-5 in rat mesenteric small
arteries. Am J Physiol Heart Circ Physiol, 294, H961-9.
Szuster-Ciesielska A, Plewka K, Daniluk J (2011). Betulin and
betulinic acid attenuate ethanol-induced liver stellate cell
activation by inhibiting reactive oxygen species (ROS),
cytokine (TNF-alpha, TGF-beta) production and by
inuencing intracellular signaling. Toxicology, 280, 152-63.
Yan G, Graham K, Lanza-Jacoby S (2012). Curcumin enhances
the anticancer effects of trichostatin a in breast cancer cells.
Mol Carcinog.
Yao QY, Xu BL, Wang JY, et al (2012). Inhibition by curcumin
of multiple sites of the transforming growth factor-beta1
signalling pathway ameliorates the progression of liver
fibrosis induced by carbon tetrachloride in rats. BMC
Complement Altern Med, 12, 156.
Ye B, Jiang LL, Xu HT, et al (2012). Expression of PI3K/AKT
pathway in gastric cancer and its blockade suppresses tumor
growth and metastasis. Int J Immunopathol Pharmacol, 25,
627-36.
Yodkeeree S, Ampasavate C, Sung B, et al (2010). curcumin
suppresses migration and invasion of MDA-MB-231
humanbreast cancer cell line. Eur J Pharmacol, 627, 8-15.
Zayani Y, Allal-Elasmi M, Jacob MP, et al (2012).Abnormal
circulating levels of matrix metalloprotei- nases and their
inhibitors in diabetes mellitus. Clin La, 58, 779-85.
Zhang SS, Gong ZJ, Li WH, et al (2012). Antibrotic effect of
curcumin in TGF-beta 1-induced myobroblasts from human
oral mucosa. Asian Pac J Cancer Pre, 13, 289-94.
Zhang YE (2009). Non-Smad pathways in TGF-beta signaling.
Cell Res, 19, 128-39.
... TGF-β cooperates with SCs to promote axon growth (Mo et al., 2012). SCs at the site of nerve injury function as a "bridge" to guide the extension and joining of severed nerve endings. ...
... This process requires the participation of various cofactors. MMPs degrade and remodel the ECM (Mo et al., 2012). However, it was recently demonstrated that MMP9 knockdown and overexpression in cultured rat SCs inhibited and promoted cell migration, respectively . ...
Article
Full-text available
Injuries caused by trauma and neurodegenerative diseases can damage the peripheral nervous system and cause functional deficits. Unlike in the central nervous system, damaged axons in peripheral nerves can be induced to regenerate in response to intrinsic cues after reprogramming or in a growth-promoting microenvironment created by Schwann cells. However, axon regeneration and repair do not automatically result in the restoration of function, which is the ultimate therapeutic goal but also a major clinical challenge. Transforming growth factor (TGF) is a multifunctional cytokine that regulates various biological processes including tissue repair, embryo development, and cell growth and differentiation. There is accumulating evidence that TGF-β family proteins participate in peripheral nerve repair through various factors and signaling pathways by regulating the growth and transformation of Schwann cells; recruiting specific immune cells; controlling the permeability of the blood-nerve barrier, thereby stimulating axon growth; and inhibiting remyelination of regenerated axons. TGF-β has been applied to the treatment of peripheral nerve injury in animal models. In this context, we review the functions of TGF-β in peripheral nerve regeneration and potential clinical applications.
... Increased expression of TGF-β enhances the inflammatory process, fibrosis, subsequent proliferation, and growth of tumor cells [100]. Thus, overproduction of TGF-β induces resistance of cancer cells into chemotherapy [101]. Similarly, TGF-β can stimulate epithelial-mesenchymal transition (EMT) via the upregulation of Smad2/3 [102]. ...
... SIRT1, a member of the mammalian sirtuin protein family, is significantly involved in several biological processes, including DNA repair, gene silencing, cell survival, metabolism, and aging [99]. Previously, the role of SIRT1 in fibrosis was supported in several organs such as the liver, heart, and kidneys [100,101]. ...
Article
Full-text available
In cancer management, drug resistance remains a challenge that reduces the effectiveness of chemotherapy. Several studies have shown that curcumin resensitizes cancer cells to chemotherapeutic drugs to overcome resistance. In the present study, we investigate the potential therapeutic role of curcumin in regulating the proliferation of drug-resistant cancers. Six drug-sensitive (MCF7, HCT116, and A549) and -resistant (MCF7/TH, HCT116R, and A549/ADR) cancer cell lines were treated with curcumin followed by an analysis of cytotoxicity, LDH enzyme, total reactive oxygen species, antioxidant enzymes (SOD and CAT), fibrosis markers (TGF-β1 protein, fibronectin, and hydroxyproline), and expression of cellular apoptotic markers (Bcl-2, Bax, Bax/Bcl-2 ratio, Annexin V, cytochrome c, and caspase-8). Additionally, the expression of cellular SIRT1 was estimated by ELISA and RT-PCR analysis. Curcumin treatment at doses of 2.7–54.3 µM significantly reduced the growth of sensitive and resistant cells as supported with decreased viability and increased cellular LDH enzyme of treated cells compared to controls non-treated cells. Curcumin also at doses of 2.7 and 54.3 µM regulated the fibrogenesis by reducing the expression of fibrotic markers in treated cells. Analysis of apoptotic markers indicated increased Bax, Bax, Bax/Bcl-2 ratio, Annexin V, caspase-8, and cytochrome c expression, while Bcl-2 expressions were significantly reduced. In curcumin-treated cells at 2.7 μM, non-significant change in ROS with significant increase in SOD and CAT activity was observed, whereas an increase in ROS with a reduction in respective antioxidant enzymes were seen at higher concentrations along with significant upregulation of SIRT1. In conclusion, the present study shows that curcumin induces anticancer activity against resistant cancer cell lines in a concentration- and time-dependent manner. The protective activities of curcumin against the growth of cancer cells are mediated by modulating oxidative stress, regulating fibrosis, SIRT1 activation, and inducing cellular apoptosis. Therefore, curcumin could be tested as an auxiliary therapeutic agent to improve the prognosis in patients with resistant cancers.
... CUR possesses a wide range of pharmacological activities and has been extensively studied for new drug development. Furthermore, the coadministration of CUR with other phytochemicals improves anticancer activity by regulating multiple molecular targets [14] and modulating various intracellular signaling pathways, including transcription factors such as NF-κB [15], Akt [16], CDK, MAPK [17], ERK [18], and Bcl-2 [19]. In traditional Chinese medicine, curcumin was employed in clinical meta-analyses for the treatment of conditions such as arthritis [20], pain and analgesia [21], menstrual pattern disorders [22], and premenstrual syndrome [23]. ...
Article
Full-text available
Breast cancer (BC) has become the fifth most prevalent cause of cancer-related morbidity, attracting significant attention from researchers due to its heightened malignancy and drug resistance. Conventional chemotherapy approaches have proven inadequate in addressing all BC subtypes, highlighting the urgent need for novel therapeutic approaches or drugs. Curcumin (CUR), a phytochemical derived from Curcuma longa (turmeric), has shown substantial potential in inhibiting BC cell migration, metastasis, and proliferation. However, the use of CUR in this context comes with challenges due to its dynamic and easily degradable nature, poor aqueous solubility, low bioavailability, rapid metabolism, and swift systemic elimination, collectively limiting its clinical applications. As such, we provide an overview of the properties, synthesis, and characterization of the hybridization of CUR and its analogue with chemo-drug building blocks. We reviewed research from the last five years on CUR’s biogenesis with respect to the regulation of BC, revealing that CUR participates in arresting BC cells in the cell cycle and significantly induces apoptosis in BC cells. Information on the chemotherapeutic and antitumor mechanisms of CUR in BC, including regulation of the cell cycle, increased cell apoptosis, and inhibition of multidrug resistance (MDR), was compiled. Additionally, we provide an overview of CUR loaded into nanomaterials that are cotreated with other chemotherapeutic drugs, such as paclitaxel, thymoquinone, and tamoxifen. In this review, we discuss different types of nanoparticles that can be used for CUR delivery, such as polymeric nanoparticles, carbon nanotubes, and liposomes. By comparing the size, entrapment efficiency, drug-loading capacity, release time, biocompatibility, pharmaceutical scale, and reproducibility of various nanomaterials, we aimed to determine which formulations are better suited for loading CUR or its analogue. Ultimately, this review is expected to offer inspiring ideas, promising strategies, and potential pathways for developing advanced anti-BC strategy nanosystems in clinical practice.
... In MDA-MB-231 cells, curcumin substantially reduced TGF-1-induced phosphorylation of Smad2, ERK1/2, and P38MAPK at the protein level as well as induced MMP-9 protein expression inhibited. The inhibition impact was concentration-dependent, with a dose of 10 µM being considered to have the strongest inhibitory effects [124]. ...
Article
The uncontrolled growth and spread of aberrant cells characterize the group of disorders known as cancer. According to GLOBOCAN 2022 analysis of cancer patients in either developed countries or developing countries the main concern cancers are breast cancer, lung cancer, and liver cancer which may rise eventually. Natural substances with dietary origins have gained interest for their low toxicity, anti-inflammatory, and antioxidant effects. The evaluation of dietary natural products as chemopreventive and therapeutic agents, the identification, characterization, and synthesis of their active components, as well as the enhancement of their delivery and bioavailability, have all received significant attention. Thus, the treatment strategy for concerning cancers must be significantly evaluated and may include the use of phytochemicals in daily lifestyle. In the present perspective, we discussed one of the potent phytochemicals, that has been used over the past few decades known as curcumin as a panacea drug of the "Cure-all" therapy concept. In our review firstly we included exhausted data from in-vivo and in-vitro studies on breast cancer, lung cancer, and liver cancer which act through various cancer-targeting pathways at the molecular level. Now, the second is the active constituent of turmeric known as curcumin and its derivatives are enlisted with their targeted protein in the molecular docking studies, which help the researchers design and synthesize new curcumin derivatives with respective implicated molecular and cellular activity. However, curcumin and its substituted derivatives still need to be investigated with unknown targeting mechanism studies in depth.
... MMP9 can degrade basement membrane collagens, leading tumor cells to invade, migrate and metastasize [22]. Consistent with our results, Na Mo and colleagues have previously reported that exposure to Curcumin inhibited breast cancer cell invasion through downregulation of TGF-β1-stimulated MMP-9 expression [23]. Furthermore, it was illustrated that endosomal Curcumin could reduce MMP9 expression and inhibit invasive tumor appearance in breast cancer mouse models [24]. ...
Article
Background: Despite the attractive anti-cancer effects, poor solubility and low bioavailability have restricted the clinical application of Curcumin. Recent findings show that Gemini nano-curcumin (Gemini-Cur) significantly improves the cellular uptake of Curcumin and its anti-cancer effect in tumor cells. Here, we aimed to assess the suppressive effect of Gemini-Cur on 4T1 breast cancer cells in vitro and, subsequently, in BALB/c mouse models. Materials and methods: Fluorescence microscopy was employed to visualize cellular uptake and morphological changes of 4T1 cells during treatment with Gemini-Cur and void curcumin. MTT and annexin V/FITC assays were performed to study the toxic effect of Gemini-Cur on mouse cancer cells. For in vivo studies, BALB/c tumor-bearing mice were used to evaluate the inhibitory effect of Gemini-Cur in comparison with mice receiving free Curcumin and nanoparticles. Results: Our data showed that Gemini-Cur enters the cells and inhibits proliferation in a time- and dose-dependent manner. Annexin V/FITC confirmed apoptotic effect on 4T1 cells. In vivo studies also illustrated that tumor growth is suppressed in Gemini-Cur treated mice rather than controls. Expression studies demonstrated the modulation of apoptotic and metastatic genes, including Bax, Bcl-2, MMP-9, VEGF, and COX-2 in treated mice. Conclusion: In conclusion, these data demonstrate the promising anti-cancer properties of Gemini-Cur on mice models. However, further studies at molecular and cellular levels are required to conclude this therapeutic advantage.
... In Epidermal Growth Factor (EGF) stimulated HT-1080 cells, capsaicin decreases the expression of MMP-9, MMP-2, and MT1-MMP through FAK/Akt, PKC/Raf/ERK, P 38 MAPK and AP-1 signaling pathway [111]. In TGF-β (10 nM) induced human breast cancer cell lines, MDA-MB-231, curcumin (≤10 μM) decreases MMP-9 protein expression [112]. Human aortic smooth muscle cells (HASMCs) treated with TNF-α increase the expression of MMP-9. ...
Article
Immunologists have long considered inflammation to be a two-edged sword. Short term inflammation can be beneficial but long term chronic inflammation is damaging. Obesity, type 2 diabetes (T2D), and cancer have recently been added to the never-ending list of inflammatory diseases. The nuclear transcription factor peroxisome proliferator-activated receptor gamma (PPAR-γ) is involved in inflammation and obesity. Clinicians employed PPAR-γ agonists, both synthetic and natural, to treat disorders such as obesity and T2D without fully understanding the biochemical features and potential adverse effects. This is one of the reasons for the controversy surrounding the thiazolidinedione class of medicines, including rosiglitazone and pioglitazone.
... Gold et al. reported curcumin and emodin down-regulated TGF-β signaling in cervical cancer cells [40]. Evidence suggests that curcumin-induced apoptosis and reversed EMT through the downregulation of the TGF-β-signaling cascade in pancreatic cancer [41] and breast cancer [42]. In our wound healing assay, combined curcumin and luteolin synergistically inhibit migration and invasion in vitro. ...
Article
Full-text available
Simple Summary One of the significant issues of the anti-cancer effects of phytochemicals, bioactive compounds from foods, and other plants, is that the effective dosages of the phytochemicals are too high to be obtained by oral intake, particularly by food intake. The current study aimed to assess if the combination of two phytochemicals, luteolin (LUT) and curcumin (CUR), at low dosages where LUT or CUR alone has no significant effect, synergistically exerts anti-colon cancer. Our results show that combined LUT and CUR synergistically suppressed colon cancer in cultured cells and cell-derived xenograft mice, which may be associated with two possible molecular pathways. This study provides a practical approach to treating or preventing colon cancer in humans by consuming foods having high levels of luteolin and curcumin. Abstract This study aimed to select a combination of curcumin and luteolin, two phytochemicals from food, at lower concentrations with a higher inhibitory effect on colon cancer growth and investigate possible molecular mechanisms of this anti-colon cancer effect. By pairwise combination screening, we identified that the combination of curcumin (CUR) at 15 μM and luteolin (LUT) at 30 μM (C15L30) synergistically suppressed the proliferation of human colon cancer CL-188 cells, but the individual chemicals had a little inhibitory effect at the selected concentrations. This result was also confirmed in other colon cancer DLD-1cells, suggesting that this synergistic inhibitory effect of C15L30 applies to different colon cancer cells. The combination C15L30 synergistically suppressed the wound closure (wound healing assay) in CL-188 cells. We also found that the combination of CUR and LUT (at 20 mg/kg/day and 10 mg/kg/day, respectively, IP injection, 5 days for 2 weeks) synergistically suppressed tumor growth in CL-188 cell-derived xenograft mice. Western blot results showed that protein levels of Notch1 and TGF-β were synergistically reduced by the combination, both in CL-188 cells and xenograft tumors. Tumor pathological analysis revealed that combined CUR and LUT synergistically increased necrosis, but the individual treatment with CUR and LUT had no significant effect on tumor necrosis. Therefore, combined curcumin and luteolin synergically inhibit colon cancer development by suppressing cell proliferation, necrosis, and migration associated with Notch1 and TGF-β pathways. This study provides evidence that colon cancer may be prevented/treated by consuming foods having high levels of luteolin and curcumin in humans.
... MMP9 is capable to degrade basement membrane collagens, leading tumor cells to invade, migrate and metastasize [18]. Consistent with our results, Na Mo and colleagues have previously reported that exposure to curcumin inhibited breast cancer cell invasion through downregulation of TGF-β1-stimulated MMP-9 expression [19]. Furthermore, it was illustrated that endosomal curcumin could reduce MMP9 expression and inhibit invasive tumor appearance in breast cancer mice models [20]. ...
Preprint
Full-text available
Background/aim Despite the attractive anticancer effects, poor solubility and low bioavailability have restricted the clinical application of curcumin. Recent findings show that gemini nano-curcumin (Gemini-Cur) significantly improves cellular uptake of curcumin and its anticancer effect in tumor cells. Here, we aimed to assess the suppressive effect of Gemini-Cur on 4T1 breast cancer cells in vitro and, subsequentlly on BALB/c mouse models. Materials and methods Fluorescence microscopy was employed to visualize cellular uptake and morphological changes of 4T1 cells during treatement with Gemini-Cur and void curcumin. MTT and annexin V/FITC assays were performed to study the toxic effect of Gemini-Cur on mouse cancer cells. For in vivo studies, BALB/c tumor-bearing mice were used to evaluate the inhibitory effect of Gemini-Cur in comparison with mice receiving free curcumin and nanoparticles. Results Our data showed that Gemini-Cur enters the cells and inhibits proliferation in a time- and dose-dependent manner. Annexin V/FITC confirmed apoptotic effect on 4T1 cells. In vivo studies also illustrated that tumor growth is suppressed in Gemini-Cur treated mice rather than controls. Expression studies demonstrated the modulation of apoptotic and metastatic genes including Bax, Bcl-2, MMP-9, VEGF and COX-2 in treated mice. Discussion Taken together, these data demonstrate the promising anticancer properties of Gemini-Cur on mice models. However, further studies in molecular and cellular levels are required to conclude this therapeutic advantage.
Chapter
Breast cancer is the most commonly diagnosed but one of the aggressive cancers among the females worldwide. The currently employed treatments for breast cancers are invasive, painful and suffer from high costs. Plant kingdom is a rich source of diverse groups of therapeutic compounds and drugs which can serve as cost effective and less toxic alternative for the breast cancer therapy. Traditionally, Chinese Traditional Medicine (CTM) and Ayurveda systems have mention of effectiveness of many herbal drugs and formulations in breast cancer therapy. The preclinical studies have pointed out towards less explored potential of phytocompounds in breast cancer chemoprevention and treatment. Plant derived compounds exhibit strong antioxidant, anti-inflammatory, immuno-modulatory and anti-angiogenic activities which may be efficiently targeted for breast cancer therapy.
Article
Full-text available
Background: Breast cancer is the most common cancer of women and the principal cause of death in middle aged women. The objective of this study was to describe the trend of breast cancer and its management in Aden and adjacent south-eastern governorates of Yemen during the last 20 years. Patients and methods: This is a retrospective analysis of previous studies on patients with breast cancer in Aden and adjacent south-eastern governorates, Yemen (January 1989 through December 2007). The studied variables were: sex, age, time and type of presentation, disease stage, pathological types and the performed surgical treatment. The sources of information were the treatment registry of Aden health office, archives of Al-Gamhouria teaching hospital; major referral and other public and private hospitals in Aden and Aden Cancer Registry. Results: The total number of patients was 476, 99% being females. The age range was 19-88 years. The most affected age was 30-50 years (60.5%), 95% presenting after one month of having breast symptoms. Forty-five percent presented with signs of advanced local disease, while 59.2% had palpable axillary lymph nodes on presentation. Early breast cancer (stages I-II) occurred in 47%, and late breast cancer (stages III-IV) in 51.5%. Invasive ductal carcinoma was the commonest pathology (89.3%). The main surgical treatment was mastectomy (modified radical mastectomy (50%). Conclusion: Breast cancer is predominantly a disease of young with late presentation and advanced disease. Improving health awareness and earlier diagnosis of the disease by health education, encouraging breast self-examination, and providing the mammography equipment and mammary clinics in hospitals are recommended. Establishment of oncology and radiotherapy centers in Aden is a necessity.
Article
Full-text available
Smooth muscle cells are exposed to growth factors and cytokines that contribute to pathological states including airway hyperresponsiveness, atherosclerosis, angiogenesis, smooth muscle hypertrophy, and hyperplasia. A common feature of several of these conditions is migration of smooth muscle beyond the initial boundary of the organ. Signal transduction pathways activated by extracellular signals that instigate migration are mostly undefined in smooth muscles. We measured migration of cultured tracheal myocytes in response to platelet-derived growth factor, interleukin-1β, and transforming growth factor-β. Cellular migration was blocked by SB203580, an inhibitor of p38MAPK. Time course experiments demonstrated increased phosphorylation of p38MAPK. Activation of p38MAPK resulted in the phosphorylation of HSP27 (heat shockprotein 27), which may modulate F-actin polymerization. Inhibition of p38MAPK activity inhibited phosphorylation of HSP27. Adenovirus-mediated expression of activated mutant MAPK kinase 6b(E), an upstream activator for p38MAPK, increased cell migration, whereas overexpression of p38α MAPK dominant negative mutant and an HSP27 phosphorylation mutant blocked cell migration completely. The results indicate that activation of the p38MAPK pathway by growth factors and proinflammatory cytokines regulates smooth muscle cell migration and may contribute to pathological states involving smooth muscle dysfunction.
Article
Full-text available
Phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway plays a crucial role in the formation and progression of many malignancies, and has been shown to be an important therapeutic target for cancer. In the present study, human gastric adenocarcinoma tissues of different grades (N=45) were collected. The protein expression of PI3Kp85alpha and phosphorylated AKT (p-AKT) was evaluated immunohistochemically in the biopsy samples. PI3K/AKT pathway was blocked by constructed recombinant small hairpin RNA adenovirus vector rAd5-PI3Kp85alpha (rAd5-P) used to transfect into human gastric cancer SGC-7901cell line. The transfection efficiency of rAd5-P in SGC-7901 cells was observed under fluorescent microscope. The expression of PI3Kp85alpha, p-AKT, Ki-67 and matrix metallopeptidase-2 (MMP-2) was detected by Real-time PCR and Western blot assays. Cell proliferative activities and metastatic capabilities were determined by MTT and Transwell assays. As a consequence, the protein expression of PI3Kp85alpha and p-AKT was respectively observed in 80.0 percent and 82.2 percent gastric adenocarcinoma tissues, elevating with the ascending order of tumor malignancy. Targeted blockade of PI3K pathway decreased the expression of PI3Kp85alpha, p-AKT, Ki-67 and MMP-2, and inhibited the proliferative activities and metastatic capabilities of gastric cancer cells. In conclusion, PI3Kp85alpha and p-AKT were strongly expressed in gastric adenocarcinoma tissues, and targeted blockade of PI3K pathway may inhibit gastric cancer growth and metastasis through down-regulation of Ki-67 and MMP-2 expression. PI3K/AKT pathway may represent an important therapeutic target for gastric cancer.
Article
Full-text available
Curcumin (CM) possesses anti-cancer activity against a variety of tumors. Matrix metalloproteinases (MMPs) play an important role in remodeling the extracellular matrix and their activities are regulated by tissue inhibitor of metalloproteinases (TIMPs) family. Control of MMP and TIMP activity are now of great significance. In this study, the effect of CM is investigated on metastatic MMPs and anti-metastatic TIMPs genes on MDA breast cancer cells cultured in a mixture of DMEM and Ham's F12 medium and treated with different concentrations of CM (10, 20 and 40μM for various lengths of time. Reverse transcription followed by quantitative real time PCR was used to detect the gene expression levels of MMPs and TIMPs in CM-treated versus untreated cases and the data were analyzed by one-way ANOVA. At high concentrations of curcumin, TIMP-1, -2, -3 and -4 genes were up-regulated after 48 hours of treatment, their over-expression being accompanied by down-regulation of MMP-2 and MMP-9 gene expression levels in a concentration- and time-dependent manner. These results suggest that curcumin plays a role in regulating cell metastasis by inhibiting MMP-2 and MMP-9 and up-regulating TIMP1 and TIMP4 gene expression in breast cancer cells.
Article
Full-text available
Background At present there is no effective and accepted therapy for hepatic fibrosis. Transforming growth factor (TGF)-β1 signaling pathway contributes greatly to hepatic fibrosis. Reducing TGF-β synthesis or inhibiting components of its complex signaling pathway represent important therapeutic targets. The aim of the study was to investigate the effect of curcumin on liver fibrosis and whether curcumin attenuates the TGF-β1 signaling pathway. Methods Sprague–Dawley rat was induced liver fibrosis by carbon tetrachloride (CCl4) for six weeks together with or without curcumin, and hepatic histopathology and collagen content were employed to quantify liver necro-inflammation and fibrosis. Moreover, the mRNA and protein expression levels of TGF-β1, Smad2, phosphorylated Smad2, Smad3, Smad7 and connective tissue growth factor (CTGF) were determined by quantitative real time-PCR, Western blot, or immunohistochemistry. Results Rats treated with curcumin improved liver necro-inflammation, and reduced liver fibrosis in association with decreased α-smooth muscle actin expression, and decreased collagen deposition. Furthermore, curcumin significantly attenuated expressions of TGFβ1, Smad2, phosphorylated Smad2, Smad3, and CTGF and induced expression of the Smad7. Conclusions Curcumin significantly attenuated the severity of CCl4-induced liver inflammation and fibrosis through inhibition of TGF-β1/Smad signalling pathway and CTGF expression. These data suggest that curcumin might be an effective antifibrotic drug in the prevention of liver disease progression.
Article
Background: The aim of this study was to determine plasma levels of matrix metalloproteinases (MMPs) 2, 3, and 9 and their tissue inhibitors (TIMPs) 1 and 2 in type 2 diabetic patients (T2DM) compared to healthy subjects. Methods: The study involved 54 patients with T2DM and 57 age and gender matched healthy adults as controls. MMPs 2 and 9 were analyzed by gelatin zymography and MMP-3 and TIMPs 1 and 2 by ELISA. Results: For technical feasibility, MMPs 2 and 9 were expressed in pro forms. Pro-MMP-9 was significantly higher (p < 0.05), whereas TIMP-1 and TIMP-2 levels were significantly decreased (p < 0.01) in patients with T2DM compared to controls. The MMP-3/TIMP-1 and the MMP-3/TMP-2 ratios were significantly higher in T2DM patients than controls (p < 0.05). Fasting plasma glucose was inversely correlated with TIMP-1 (r = -0.412, p < 0.01) and TIMP-2 (r = -0.315, p < 0.001), but was not associated with MMPs. Conclusions: The present study identified abnormalities in plasma markers for extracellular matrix metabolism in T2DM. The new parameters would constitute an effective approach to explore the complications of uncontrolled diabetes.
Article
Cited (CBP/p300-interacting transactivators with glutamic acid (E)/aspartic acid (D)-rich C-terminal domain) 2, which is a CBP/p300-binding transcription co-activator without typical DNA-binding domains, has been implicated in control of cell growth and malignant transformation in Rat1 cells. In this report, we provide evidence that Cited2 is an important regulator of transforming growth factor (TGF)-β signaling. Overexpression of Cited2 enhanced TGF-β-mediated transcription of a Smad-Binding Element-containing luciferase reporter construct, SBE4-Luc. This may occur through a direct physical association of Cited2 with Smads 2 and 3, as supported by co-immunoprecipitation, mammalian two-hybrid and glutathione S-transferase-pull down assays. The transcription factor p300, which binds to Smad3, was shown to further enhance the interaction between Cited2 and Smad3, and the transcriptional responses of Smad3 by Cited2 in reporter assays. Cited2 enhances TGF-β-mediated upregulation of matrix metalloproteinase 9 (MMP9) in Cited2 inducible mouse embryo fibroblasts. Overexpression of Cited2 enhanced TGF-β-mediated MMP9 promoter reporter activity. Moreover, knockdown of Cited2 in MDA-MB-231 cells attenuated TGF-β-mediated upregulation of MMP9 and TGF-β-mediated cell invasion. Chromatin immunoprecipitation showed that Cited2 and Smad3 were recruited to MMP9 promoter upon TGF-β stimulation. This is the first demonstration that Cited2 functions as a Smad3/p300-interacting transcriptional co-activator in modulating the expression of MMP9, which could affect tumor cell invasion mediated by TGF-β.
Article
TGF-β is a ubiquitous cytokine that plays an active role in many cellular processes. Nearly every cell type has the ability to secrete TGF-β, as well as the ability to respond to TGF-β via the presence of TGF-β receptors on the cell surface. Consequently, gain or loss of function of the TGF-β pathway and its components are known to lead to a variety of diseases, including cancer. In epithelial cells, TGF-β functions as a tumor suppressor, where it inhibits proliferation, induces apoptosis, and mediates differentiation. Conversely, in other contexts, TGF-β promotes tumor progression through increasing tumor cell invasion and metastasis. Thus, TGF-β can have opposing roles, likely dependent, in part, on whether the cancer is early or late stage. The effects of TGF-β on tumor suppression and promotion are not limited to the tumor cell itself; rather, these effects can also be mediated through the stroma and the immune system. The dichotomous role of TGF-β in cancer highlights our need to understand the contextual effects of this cytokine to better guide patient selection for the use of anti-TGF-β therapies currently in clinical trials.
Article
Myofibroblasts play an important role in the development of oral submucous fibrosis (OSF). In the current study, we investigate the effect of curcumin on growth and apoptosis of myofibroblasts derived from human oral mucosa. Myofibroblasts were generated by incubating fibroblasts, obtained from human oral mucosa, with transforming growth factor-β 1 (TGF-β 1). MTT, PI staining, and FACS assays were used to investigate curcumin's effect on proliferation and cell cycle of fibroblasts and myofibroblasts. Annexin V/PI binding and FACS assays were used to examine apoptosis of myofibroblasts, Western blotting to determine the levels of Bcl-2 and Bax, and enzyme-linked immunosorbant assay was employed to examine the levels of collagen type I and III in the supernatants of myofibroblasts. Curcumin inhibits proliferation of fibroblasts and myofibroblasts; it also disturbs the cell cycle, induces apoptosis and decreases the generation of collagen type I and III in myofibroblasts, which are more sensitive to its effects than fibroblasts. Curcumin induces apoptosis in myofibroblasts by down-regulating the Bcl-2/ Bax ratio. Our results demonstrate the antifibrotic effect of curcumin in vitro. It may therefore be a candidate for the treatment of OSF.