ArticlePDF Available

Kaempferol Inhibits Angiogenesis and VEGF Expression Through Both HIF Dependent and Independent Pathways in Human Ovarian Cancer Cells

Taylor & Francis
Nutrition and Cancer
Authors:

Abstract and Figures

Ovarian cancer is 1 of the most significant malignancies in the Western world, and the antiangiogenesis strategy has been postulated for prevention and treatment of ovarian cancers. Kaempferol is a natural flavonoid present in many fruits and vegetables. The antiangiogenesis potential of kaempferol and its underlying mechanisms were investigated in two ovarian cancer cell lines, OVCAR-3 and A2780/CP70. Kaempferol mildly inhibits cell viability but significantly reduces VEGF gene expression at mRNA and protein levels in both ovarian cancer cell lines. In chorioallantoic membranes of chicken embryos, kaempferol significantly inhibits OVCAR-3-induced angiogenesis and tumor growth. HIF-1alpha, a regulator of VEGF, is downregulated by kaempferol treatment in both ovarian cancer cell lines. Kaempferol also represses AKT phosphorylation dose dependently at 5 to 20 muM concentrations. ESRRA is a HIF-independent VEGF regulator, and it is also downregulated by kaempferol in a dose-dependent manner. Overall, this study demonstrated that kaempferol is low in cytotoxicity but inhibits angiogenesis and VEGF expression in human ovarian cancer cells through both HIF-dependent (Akt/HIF) and HIF-independent (ESRRA) pathways and deserves further studies for possible application in angio prevention and treatment of ovarian cancers.
Content may be subject to copyright.
Kaempferol Inhibits Angiogenesis and VEGF Expression Through
Both HIF Dependent and Independent Pathways in Human Ovarian
Cancer Cells
Haitao Luo,
Alderson-Broaddus College, Philippi, West Virginia, USA
Gary O. Rankin,
Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
Lingzhi Liu,
Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia, USA
Matthew K. Daddysman,
Alderson-Broaddus College, Philippi, West Virginia, USA
Bing-Hua Jiang, and
Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia, USA
Yi Charlie Chen
Alderson-Broaddus College, Philippi, West Virginia, USA
Abstract
Ovarian cancer is 1 of the most significant malignancies in the Western world, and the
antiangiogenesis strategy has been postulated for prevention and treatment of ovarian cancers.
Kaempferol is a natural flavonoid present in many fruits and vegetables. The antiangiogenesis
potential of kaempferol and its underlying mechanisms were investigated in two ovarian cancer cell
lines, OVCAR-3 and A2780/CP70. Kaempferol mildly inhibits cell viability but significantly reduces
VEGF gene expression at mRNA and protein levels in both ovarian cancer cell lines. In
chorioallantoic membranes of chicken embryos, kaempferol significantly inhibits OVCAR-3-
induced angiogenesis and tumor growth. HIF-1α, a regulator of VEGF, is downregulated by
kaempferol treatment in both ovarian cancer cell lines. Kaempferol also represses AKT
phosphorylation dose dependently at 5 to 20 μM concentrations. ESRRA is a HIF-independent VEGF
regulator, and it is also downregulated by kaempferol in a dose-dependent manner. Overall, this study
demonstrated that kaempferol is low in cytotoxicity but inhibits angiogenesis and VEGF expression
in human ovarian cancer cells through both HIF-dependent (Akt/HIF) and HIF-independent
(ESRRA) pathways and deserves further studies for possible application in angio prevention and
treatment of ovarian cancers.
INTRODUCTION
Ovarian cancer is one of the most important malignancies for women in the Western world,
ranking as the fifth leading cause of cancer-related deaths (1). Due to a lack of effective
Copyright © 2009, Taylor & Francis Group, LLC
Address correspondence to Yi Charlie Chen, Natural Science Division, Alderson-Broaddus College, 101 College Hill Drive, Philippi,
WV 26416. Phone: 304-457-6277. Fax: 304-457-6239. chen@ab.edu.
NIH Public Access
Author Manuscript
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
Published in final edited form as:
Nutr Cancer. 2009 ; 61(4): 554–563. doi:10.1080/01635580802666281.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
biomarkers for screening, nearly 60–70% of ovarian cancers are diagnosed at advanced stages
(2), with a poor prognosis of about 30% for a 5-yr survival rate (3). Treatment of ovarian
cancers involves surgery and chemotherapy, but is often not effective because of problems
with drug resistance (4) and later relapse in patients (5).
Angiogenesis is the process of developing new blood vessels and plays an important role in
tumor growth (6). In a normal adult, angiogenesis is virtually quiescent, with only 0.01% of
endothelial cells undergoing cell division (7). In contrast, tumor growth requires active
angiogenesis, and antiangiogenesis becomes a rational anticancer strategy (7). Vascular
endothelial growth factor (VEGF) is the most pivotal positive regulator of angiogenesis (6),
and VEGF gene expression is found in many human tumors including ovarian cancers (8).
VEGF gene expression is regulated by oxygen tension, growth factors, hormones, and
oncogenes (9). Hypoxia induces VEGF expression through hypoxia-inducible factor 1 (HIF-1),
which is composed of HIF-1α and HIF-1β subunits, with the former one being inducible (10).
In normoxia, the PI3 kinase/AKT pathway is implicated in the regulation of HIF-mediated
VEGF responses (11). Growth factors and inflammatory cytokines, including epidermal
growth factor, transforming growth factor (TGF), interleukin-1 (IL-1), IL-6, also stimulate
expression of the VEGF gene (9). Estrogens activate VEGF expression through estrogen
receptors (ERs) and the estrogen response element (ERE) (12). Proto-oncogene c-Myc enforces
cellular proliferation and growth in tumors (13) and cooperates with HIF-1 in inducing VEGF
expression (14). Myc has been recently reported to be regulated by PI3K/AKT pathways
(15). Whereas regulation of VEGF through PI3Kinase/AKT and HIF is considered the classical
pathway, a pathway involving peroxisome proliferator-activated receptor gamma coactivator
1 alpha (PPARGC1A) and estrogen-related receptor alpha (ESRRA) was recently discovered
to be independent of HIF (16). This pathway goes through ESRRA, an orphan nuclear receptor
that has a high degree of sequence similarity and intense cross-talk to ERs (17).
Flavonoids are natural polyphenols present in a wide variety of fruits and vegetables (19) and
have been shown to inhibit cancer development (20,21). It has been reported that dietary
flavonoids reduce the risk to cardiovascular disease (22), prostate cancer (23), colorectal cancer
(24), and renal cancer (19) in humans. Flavonoids have also been found to inhibit cell growth
and proliferation (25) and induce cell toxicity (26,27) in cancer cells. Kaempferol [3,5,7-
trihydroxy-2-(4-hydroxyphenyl)-4H-1-benzopyran-4-one] is among the most common dietary
flavonoids and recently has been shown to possess antioxidant and antitumor properties.
Kaempferol was found to inhibit proliferation of human breast and lung cancer cells (28,29),
inhibit ER-α expression in breast cancer cells (28), and induce apoptosis in glioblastoma cells
(30) and in lung cancer cells by activation of MEK-MAPK (29). Kaempferol also exhibited an
anti-inflammatory effect through inhibition of IL-4 (31), COX-2 and CRP expression and
downregulation of NFκ B pathway in liver cells (32). In human studies, a significant 40%
decrease in ovarian cancer incidence was found for the highest quintile of kaempferol intake
as compared to the lowest quintile (33). However, the effect and mechanism by which
kaempferol inhibits ovarian cancer cell proliferation and tumor formation is not yet clear. In
this study, kaempferol was investigated for its effects on angiogenesis and VEGF expression
in ovarian cancer cells and the underlying mechanisms for this effect, including the
conventional AKT-HIF and novel PPARGC1A-ESRRA pathways.
MATERIALS AND METHODS
Cell Culture
Two human ovarian cancer cell lines, OVCAR-3 (mutant p53) and A2780/CP70 (wild-type
p53) (34,35), were maintained in RPMI 1640 medium supplemented with 100 units/ml
penicillin, 100 μg/ml streptomycin (VWR, West Chester, PA), and 10% US-qualified fetal
Luo et al. Page 2
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
bovine serum (Invitrogen, Grand Island, NY) in a humidified incubator with 5% CO2 at 37°
C.
Cell Viability Assay
Kaempferol’s effects on OVCAR-3 and A2780/CP70 cell viability were colorimetrically
determined with a “CellTiter 96 Aqueous One Solution Cell Proliferation Assay” kit from
Promega (Madison, WI). Cells (8 × 103/well) were seeded into 96-well plates and incubated
for 16 h before being treated with 0 to 80 μM kaempferol (Sigma, St. Louis, MO) in triplicates
for another 24 h with DMSO as solvent control. Cells were then washed twice with phosphate-
buffered saline (PBS), introduced with 100 μl medium containing 3-(4,5-dimethylthiazol-2-
yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS),
incubated at 37°C for 2 h, and measured for OD values at 490 nm. A linear standard curve was
generated by seeding different amount of cells (0 to 1 × 104) and treating with medium
containing DMSO only. Cell viability was expressed as percentage of control from 3
independent experiments.
qRT-PCR
The effects of kaempferol on several genes’ mRNA level were determined by quantitative
reverse-transcription PCR (qRT-PCR). Cells (5 × 105) were seeded in 60-mm dishes and
incubated for 16 h before treatment with kaempferol. For time course of VEGF mRNA
expression, both cell lines were treated with 40 μM kaempferol for 0 to 24 h, and cells were
harvested in TRIzol reagent (Invitrogen, Grand Island, NY) and stored in 80°C until analysis.
For VEGF mRNA expression in response to kaempferol doses, both cell lines were treated
with 0 to 40 μM kaempferol for 24 h before RNA was extracted, reconstituted in DEPC-treated
water, and checked for integrity by agarose-gel electrophoresis. RNA samples were quantitated
at OD 260/280, and 1 μg RNA was introduced to reverse transcription with AMV reverse
transcriptase from Promega (Madison, WI). cDNA equivalent to 80 ng RNA was amplified by
real-time PCR for various genes in triplicate with RT2 SYBR Green qPCR Master Mix
(SuperArray, Frederick, MD) and a Chromo4™ real-time detector coupled to a DNA Engine®
thermal cycler (Bia-Rad, Hercules, CA). Primers for GAPDH, HIF-1α, HIF-1β, and ESRRA
were chosen from the Primer-Bank Web site (http://pga.mgh.harvard.edu/primerbank/), and
primers for VEGF and PPARGC1A were designed from the Primer3 Web site
(http://frodo.wi.mit.edu/cgi-bin/primer3/primer3_www.cgi). Primer sequences are listed in
Table 1. The PCR program was set as follows: 95°C 10 min (95°C 20s, 58°C 45 s, 72°C 20 s,
77°C 1 s, read plate) × 50; 72°C 5 min; 58°C 1 min; melting curve (65°C to 95°C by 0.5°C
increments). A standard curve for each gene was generated from serial dilutions of PCR
products to monitor amplification efficiency and to relatively quantify mRNA abundance.
RNA samples without reverse transcription served as a non-reverse-transcription (RT)
control, and water served as a nontemplate control (NTC). Arbitrary units of each gene were
derived from a corresponding standard curve, and mRNA abundance was normalized to
GAPDH levels and expressed as percentages of control for statistical analysis.
ELISA
Secreted VEGF protein levels were analyzed by sandwich ELISA with a Quantikine Human
VEGF Immunoassay Kit from R&D Systems (Minneapolis, MN) targeting VEGF165 in cell
culture supernates. Cells (8 × 103/well) were seeded into 96-well plates and incubated for 16
h before treatment with kaempferol for 24 h. Culture supernates were collected for VEGF
assay, and cell numbers were quantitated with MTS-based assay as mentioned above. VEGF
levels, as determined following the manufacturer’s instructions, were normalized to cell
numbers for each treatment. A total of 4 independent experiments, each in duplicates, was
assayed, and the mean VEGF protein level from each duplicate was used for statistical analysis.
Luo et al. Page 3
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
For phosphorylated ERK1/ERK2 and AKT, DuoSet IC kits (R&D Systems, Minneapolis, MN)
were used to develop sandwich ELISA, measuring phospho-ERK1 (T202/Y204)/phospho-
ERK2 (T185/Y187), and phospho-AKT (Pan) (S473) in cell lysates, respectively. Cells (5 ×
105) were seeded into 60-mm dishes and incubated for 16 h before treatment with kaempferol
for 24 h. Cell lysates were analyzed for p-ERK and p-AKT levels as per instructions, and total
protein levels in lysates were determined with BCA Protein Assay Kit (Pierce, Rockford, IL)
to normalize p-ERK and p-AKT abundance. A total of 4 independent experiments with
triplicates each were performed, and averages from each triplicate were used for statistical
analysis.
Western Blot
Cells (5 × 105) were seeded in 60-mm dishes and incubated for 16 h before treatment with
kaempferol for 24 h. After double wash with cold PBS, cells were harvested with RIPA buffer
freshly supplemented with 3 μg/ml aprotinin, 10 μg/ml leupeptin, 10 μg/ml pepstatin, 1 mM
sodium vanadate (activated), and 1 mM PMSF (Sigma, St. Louis, MO). Total protein levels
were assayed with a BCA Protein Assay Kit (Pierce, Rockford, IL), and lysates (50 μg total
protein) were separated by 10% SDS-PAGE and blotted into nitrocellulose membrane with a
Mini-Protean 3 System (Bio-Rad Laboratories, Hercules, CA). For immunodetection,
antibodies against HIF-1α, HIF-1β (BD Biosciences, San Jose, CA), and GAPDH (Santa Cruz
Biotechnology, Santa Cruz, CA) were applied and signals visualized with a SuperSignal West
Pico Complete Mouse IgG Detection Kit and x-ray film (Pierce Biotechnology, Rockford, IL).
Protein bands were quantitated with Quantity One software (Bio-Rad Laboratories, Hercules,
CA), normalized to corresponding GAPDH bands, and expressed as percentages of control. A
total of 3 independent experiments were carried out for statistical analysis.
Chicken Embryo Chorioallantoic Membrane (CAM) Assay
OVCAR-3 cells (3 × 106) were suspended in 100 μl medium (serum-free, 4°C), mixed with
50 μl liquid BD Matrigel Basement Membrane Matrix High Concentration (BD Biosciences,
San Jose, CA), supplemented with or without 20-μM kaempferol at 4°C, and implanted in the
CAM of 9-day-old chicken embryos. After 5-day incubation at 37.5°C, tumor implants were
dissected out, photographed, weighed, and counted under a dissecting microscope for
branching blood vessels. A total of 5 chicken embryos were included in each treatment group.
Statistical Analysis
Average values of replicates were collected from independent experiments and analyzed by
analysis of variance (ANOVA) and post hoc test (2-sided Dunnett’s t) with SPSS 15.0 (SPSS
Inc., Chicago, IL) to test both overall differences and specific differences between each
treatment and control. A P value of less than 0.05 was considered significant.
RESULTS
Kaempferol Mildly Inhibits Cell Viability in Ovarian Cancer Cell Lines
OVCAR-3 and A2780/CP70 cells were treated with kaempferol for 24 h and assayed for cell
viability. As shown in Fig. 1A, OVCAR-3 cell viability was inhibited to 91% by 20-μM
kaempferol treatment (P < 0.01), and to 74% at 80-μM kaempferol concentration (P < 0.001).
For A2780/CP70 cells, the viability was slightly promoted to 102% at 20-μM kaempferol
concentration (P > 0.38), and then inhibited down to 94% and 79% by 40-μM and 80-μM
kaempferol treatments, respectively (P < 0.001). An overall inhibitory effect on cell viability
was observed for both cell lines, and A2780/CP70 cells appeared more resistant than OVCAR-3
cells to the inhibiting effect of kaempferol (P < 0.05).
Luo et al. Page 4
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Kaempferol Inhibits VEGF Expression in Ovarian Cancer Cell Lines
Both cell lines were treated with kaempferol and assayed for VEGF mRNA and protein levels
by qRT-PCR and ELISA, respectively. As shown in Fig. 1B, 40-μM kaempferol treatment did
not influence VEGF mRNA expression within 8 h. However, VEGF mRNA levels were
downregulated to 57% in OVCAR-3 cells (P < 0.05) and 72% in A2780/CP70 cells (P < 0.05)
at Hour 16 and remained at close levels at Hour 24. For 24-h kaempferol treatment (Fig. 1C),
VEGF mRNA expression was downregulated by 20-μM kaempferol treatment to 73% (P <
0.05) and 81% (P < 0.001) in OVCAR-3 and A2780/CP70 cells, respectively. Significant
further downregulation was also observed for 40-μM kaempferol treatment in both cell lines.
Both cell lines showed concentration dependent inhibition on VEGF mRNA levels by
kaempferol treatment, and no significant difference between OVCAR-3 and A2780/CP70 cells
was observed.
VEGF protein levels in cell culture supernates were also downregulated by kaempferol
treatments (Fig. 1D). The levels of VEGF protein were 80% (P < 0.05) and 82% (P < 0.05) of
controls in OVCAR-3 and A2780/CP70 cells, respectively, at 10-μM kaempferol
concentration. Further inhibition was observed for higher kaempferol treatments in a
concentration-dependent and significant manner, and both cell lines appeared similar in their
response to kaempferol’s inhibition on VEGF protein levels.
Kaempferol Inhibits Tumorigenesis and Angiogenesis in Chicken Embryo CAMs
Kaempferol was tested for its effects on in vivo angiogenesis. Chicken embryo CAMs were
implanted with OVCAR-3 cells suspended in cold liquid matrigel, which gels quickly at raised
temperature. This cancer cell-containing gel becomes a tumor in chicken embryo and continues
to grow and induce angiogenesis within the vasculature-rich CAM due to the lack of an immune
system in chicken embryos. Growth and angiogenesis of tumor implants are inhibited by
inclusion of 20-μM kaempferol. As shown in Fig. 1E, the implanted cancer cells grow to a
tumor of 70 mg, with 26 blood vessels counted. Inclusion of 20-μM kaempferol in this implant,
however, reduced tumor growth down to 38 mg (P < 0.01) and inhibited blood vessel
development to 16 (P < 0.05). A typical photograph (Fig. 1E) was shown to contrast the 2
tumors with or without kaempferol in terms of both tumor size and angiogenesis.
Kaempferol Inhibits HIF-1α Protein Expression in Ovarian Cancer Cell Lines
The effect of kaempferol on HIF-1α and HIF-1β gene expression was investigated at both the
mRNA and protein levels by qRT-PCR and Western blot, respectively. As shown in Fig. 2A,
kaempferol treatment did not have an obvious effect on HIF-1α mRNA levels except for an
80-μM treatment on OVCAR-3 cells, which downregulated HIF-1α mRNA level to 80% (P =
0.010). For HIF-1β mRNA, no appreciable effects were found in both cell lines (P > 0.40),
leaving a random distribution and an inconsistent pattern (Fig. 2B).
HIF-1α protein levels showed intense and consistent down-regulation by kaempferol treatment
(Fig. 2C). A 5-μM kaempferol treatment led to inhibition of HIF-1α protein to 75% in
OVCAR-3 cells (P < 0.01), and a 10-μM treatment reduced HIF-1α protein to 70% in A2780/
CP70 cells (P < 0.05). Higher concentrations of kaempferol resulted in greater inhibition, with
the levels of HIF-1α protein at 80-μM kaempferol down to 9% and 4% in OVCAR-3 and
A2780/CP70 cancer cells, respectively (P < 0.001). For the concentration-dependent inhibition
of HIF-1α protein levels, no significant difference between OVCAR-3 and A2780/CP70 cells
was observed. HIF-1β proteins were not affected by kaempferol treatment, with levels
distributed from 91% to 118% (P > 0.50) randomly for both cell lines (Fig. 2D).
Luo et al. Page 5
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Kaempferol Inhibits p-AKT But Not p-ERK Levels in Ovarian Cancer Cell Lines
The levels of the signal transduction molecules, phosphorylated AKT and ERK, were
determined with ELISA. As shown in Fig. 3A, p-AKT levels were downregulated from 21.8
ng/mg total protein (TP) at control to 11.2 ng/mg TP at 20-μM kaempferol treatment in
OVCAR-3 cells, and from 8.8 ng/mg TP at control to 7.1 ng/mg TP at 20-μM kaempferol
treatment in A2780/CP70 cells, showing a concentration dependent and significant inhibition
in both cell lines (P < 0.05). Phosphorylation of ERK is promoted by kaempferol treatment
(Fig. 3B). The p-ERK levels were increased from 4.7 ng/mg TP at control to 6.7 ng/mg TP at
20 μM in OVCAR-3 cells (P < 0.05), showing a concentration-dependent promotion effect by
kaempferol treatment; however, kaempferol did not affect p-ERK level in A2780/CP70 cells
(P = 0.18). Both signaling molecules were found at much higher levels in OVCAR-3 cells than
in A2780/CP70 cells, and kaempferol’s effect on these signaling molecules was also more
pronounced in OVCAR-3 cells (Fig. 3).
Kaempferol Inhibits ESRRA mRNA Expression in Ovarian Cancer Cell Lines
Kaempferol’s effect on PPARGC1A and ESRRA was examined by qRT-PCR in both cell lines.
As shown in Fig. 4A, no significant change was found on PPARGC1A mRNA levels, resulting
in a random fluctuation of mRNA levels with increasing doses of kaempferol. For ESRRA,
the mRNA levels were inhibited to 62% in OVCAR-3 cells by a 20-μM treatment (P < 0.01)
and to 68% in A2780/CP70 cells at 40-μM kaempferol (P < 0.01; Fig. 4B). Further inhibition
effects were observed for higher kaempferol treatments in a concentration-dependent manner.
A2780/CP70 cells also show higher resistance than OVCAR-3 cells to kaempferol treatment
(P < 0.01), with a significant inhibition of ESRRA mRNA levels observed only at 40- and 80-
μM treatments.
DISCUSSION
The risk of ovarian cancer increases with age and with the use of oral contraceptive pills
(18). Few studies have been done to relate lifestyles to ovarian cancer risks, although ovarian
cancer risk has been consistently associated with a high intake of saturated fat and a low intake
of vegetables (36). Flavonoids, as an abundant ingredient in fruits and vegetables, are believed
to play an important role in anticarcinogenesis through their antioxidant, antiestrogenic,
antiproliferative, antiangiogenic, and anti-inflammatory properties (33). Kaempferol is a
flavonoid widely and abundantly distributed in diet, and an impressive 40% decrease in ovarian
cancer incidence with kaempferol intake reported by a recent study suggests kaempferol as a
potential chemoprevention agent but calls for more intense studies on its effect and mechanism
of action in ovarian cancers (33).
Obviously, kaempferol’s effects in reducing ovarian cancer risks can not be explained solely
for its direct cytotoxicity on ovarian cancer cells, as our experiments on ovarian cancer cell
lines only revealed a very mild inhibitory effect on cell viability. Besides, very few chemicals
can distinguish well between tumor cells and normal tissues, and a strong cytotoxicity is
normally associated with severe side effects because of the killing of healthy cells. As a widely
distributed dietary flavonoid, kaempferol does not cause any severe side effects in either
healthy people or ovarian cancer patients, nor does it kill ovarian cancer cells directly and
strongly. However, kaempferol could possibly inhibit ovarian cancer cells through an indirect
mechanism, antiangiogenesis.
Ovarian cancer cells are known to secrete VEGF to recruit vascular endothelial cells for
angiogenesis (37,38). Angiogenesis is critical in tumor growth, invasion, and metastasis (39,
40). Therefore, VEGF is one of the most significant and direct targets in an antiangiogenesis
strategy. Our experiments discovered time- and dose-dependent inhibition on VEGF
Luo et al. Page 6
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
expression in ovarian cancer cells by kaempferol, with a greater effect at protein levels than
mRNA levels. A 10-μM kaempferol treatment significantly inhibited VEGF protein secretion
down to 80%; but for VEGF mRNA, a concentration of 20 μM was needed to reach similar
VEGF inhibition. This difference possibly reflects an amplification effect in translating mRNA
to proteins in ovarian cancer cells where the signal for VEGF expression was amplified along
the central dogma in genetics. By comparing kaempferol’s inhibitory effects on cell viability
and VEGF expression, it further proves that kaempferol mainly performs its function through
antiangiogenesis rather than killing cells directly: a 10-μM kaempferol treatment inhibits
VEGF protein down to 80%, a slightly higher 20-μM kaempferol treatment inhibits VEGF
mRNA down to 80%, but a eight-fold higher concentration (80 μM) of kaempferol is needed
to inhibit cell viability down to 80% in A2780/CP70 ovarian cancer cells. In our chicken
embryo CAM assay, kaempferol also reduced tumor size significantly. This should be
explained more as a result of antiangiogenesis rather than direct inhibition of tumor growth
because a 20 μM kaempferol only inhibits OVCAR-3 cell viability down to 91% in our cell
viability assay, whereas the tumor weight was reduced down to 54% (from 70 to 38 mg).
Although our results indicate that kaempferol works on ovarian cancer cells through inhibiting
VEGF expression rather than killing cancer cells directly, inhibition of VEGF is more than just
antiangiogenesis. It, in turn, has an indirect effect in suppressing growth of cancer cells.
Originally thought to recruit vascular endothelial cells through a paracrine mode, VEGF was
later found to also act in an autocrine manner to promote growth of tumor cells themselves
(41). Ovarian tumors are known to express VEGF receptors (42), and VEGF secreted by
ovarian cancer cells can act in both autocrine and paracrine fashions to promote growth of
tumor cells. Moreover, VEGF can act as a survival factor through enhanced expression of B-
cl-2 and survivin, protecting cancer cells against apoptosis (6). By targeting VEGF in ovarian
cancer cells, kaempferol inhibits angiogenesis in tumors and represses tumor growth and
survival indirectly.
Our experiments further explored mechanisms through which kaempferol inhibits VEGF
expression and angiogenesis in ovarian cancer cells. It is known that hypoxia stimulates VEGF
expression through inducing HIF-1 transcriptional factors; and under normoxia, the PI3 kinase
and AKT signaling pathway is implicated in HIF-1-mediated responses (43). Like apigenin,
the other dietary flavonoid studied for this pathway (10), kaempferol inhibits HIF-1α protein
levels in a concentration dependent manner; but HIF-1α mRNA levels are only inhibited at
high kaempferol concentration (80 μM) in ovarian cancer cells. This again indicates an
amplification effect in the translation process. HIF-β is a constitutively expressed subunit and
as expected is not affected by kaempferol treatment at mRNA or protein levels. Phosphorylated
AKT in this pathway was downregulated by kaempferol treatment up to 20 μM in a
concentration-dependent manner, and it proves AKT-HIF-VEGF as a working mechanism at
physiologically relevant kaempferol concentrations, which is typically below 20 μM in vivo.
Normal population kaempferol intake is assumed to be the 0.8 to 11.0 mg/day in the Nurses’
Health Study (33). We also examined another signaling molecule, p-ERK, and our results
showed that ERK pathway is not involved in kaempferol’s effect on VEGF inhibition.
Independent of HIF transcription factors, the PPARGC1A/ESRRA pathway is newly
discovered that affects VEGF expression and angiogenesis (16). Kaempferol treatment has no
effect on PPARGC1A mRNA levels, but it concentration dependently repressed ESRRA
mRNA levels in ovarian cancer cells. This might be another mechanism through which
kaempferol inhibits VEGF expression Estrogens bind to nuclear receptors ERα and ERβ, which
are ligand-inducible transcription factors and stimulate transcription of many genes. Although
no classical EREs are found in the 5 regulatory regions, VEGF is an estrogen responsive gene
with several AP-1 and Sp1 sites in this region to mediate estrogens’ effect (44) and a variant
ERE 1.5 kb away from the transcription start (6). In fact, estrogens have been shown to induce
Luo et al. Page 7
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
VEGF mRNA levels in ER+ breast tumor cells (12), and kaempferol has been reported to
inhibit ERα expression at both mRNA and protein levels in breast cancer cells (28). Although
ESRRA does not respond to an estrogen stimulus, it is a downstream effector of PPARGC1A.
ESRRA is also evolutionarily related to ERs and can efficiently bind to EREs that are
commonly shared by many target genes (45). In fact, ESRRA expression has already been
suggested as a negative prognostic factor for disease-free survival of ovarian cancer patients
(17). Inhibition of ESRRA mRNA levels in ovarian cancer cells has suggested another pathway
as kaempferol’s working mechanism in antiangiogenesis.
Overall, the dietary flavonoid, kaempferol, was found to be effective in inhibiting VEGF
expression, angiogenesis, and cell viability through both the HIF-1α dependent and the
HIF-1α independent pathways in ovarian cancer cells (Fig. 5). As a widely distributed natural
product, kaempferol is low in toxicity, potent in antiangiogenesis, and deserves further study
on possible applications in angio-prevention and therapy of ovarian cancers.
Acknowledgments
This research was supported by Grant P20 RR16477 from the National Center for Research Resources awarded to the
West Virginia IDeA Network for Biomedical Research Excellence.
REFERENCES
1. Jemal A, Tiwari RC, Murray T, Ghafoor A, Samuels A, et al. Cancer statistics, 2004. CA Cancer J
Clin 2004;54:8–29. [PubMed: 14974761]
2. Fishman DA, Schwartz PE. Current approaches to diagnosis and treatment of ovarian germ cell
malignancies. Curr Opin Obstet Gynecol 1994;6:98–104. [PubMed: 8180359]
3. Greenlee RT, Hill-Harmon MB, Murray T, Thun M. Cancer statistics. CA Cancer J Clin 2001;51:15–
36. [PubMed: 11577478]
4. Perez RP, Godwin AK, Hamilton TC, Ozols RF. Ovarian cancer biology. Semin Oncol 1991;18:186–
204. [PubMed: 2042060]
5. McGuire WP, Hoskins WJ, Brady MF, Kucera PR, Partridge EE, et al. Cyclophosphamide and cisplatin
compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer. N Engl
J Med 1996;334:1–6. [PubMed: 7494563]
6. Ferrara N. Vascular endothelial growth factor as a target for anticancer therapy. Oncologist 2004;9:2–
10. [PubMed: 15178810]
7. Bertl E, Bartsch H, Gerhäuser C. Inhibition of angiogenesis and endothelial cell functions are novel
sulforaphane-mediated mechanisms in chemoprevention. Mol Cancer Ther 2006;5:575–585.
[PubMed: 16546971]
8. Hartenbach EM, Olson TA, Goswitz JJ, Mohanraj D, Twiggs LB, et al. Vascular endothelial growth
factor (VEGF) expression and survival in human epithelial ovarian carcinomas. Cancer Lett
1997;121:169–175. [PubMed: 9570355]
9. Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev
2004;25:581–611. [PubMed: 15294883]
10. Fang J, Xia C, Cao Z, Zheng JZ, Reed E, et al. Apigenin inhibits VEGF and HIF-1 expression via
PI3K/AKT/p70S6K1 and HDM2/p53 pathways. FASEB J 2005;19:342–353. [PubMed: 15746177]
11. Jiang BH, Jiang G, Zheng JZ, Lu Z, Hunter T, et al. Phosphatidylinositol 3-kinase signaling controls
levels of hypoxia-inducible factor 1. Cell Growth Differ 2001;12:363–369. [PubMed: 11457733]
12. Sengupta K, Banerjee S, Saxena N, Banerjee SK. Estradiol-induced vascular endothelial growth
factor-A expression in breast tumor cells is biphasic and regulated by estrogen receptor-alpha
dependent pathway. Int J Oncol 2003;22:609–614. [PubMed: 12579315]
13. Ray S, Atkuri KR, Deb-Basu D, Adler AS, Chang HY, et al. MYC can induce DNA breaks in vivo
and in vitro independent of reactive oxygen species. Cancer Res 2006;66:6598–6605. [PubMed:
16818632]
Luo et al. Page 8
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
14. Kim JW, Gao P, Liu YC, Semenza GL, Dang CV. Hypoxia-inducible factor 1 and dysregulated c-
Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2
and pyruvate dehydrogenase kinase 1. Mol Cell Biol 2007;27:7381–7893. [PubMed: 17785433]
15. Zhu J, Blenis J, Yuan J. Activation of PI3K/Akt and MAPK pathways regulates Myc-mediated
transcription by phosphorylating and promoting the degradation of Mad1. Proc Natl Acad Sci USA
2008;105:6584–6589. [PubMed: 18451027]
16. Arany Z, Foo SY, Ma Y, Ruas JL, Bommi-Reddy A, et al. HIF-independent regulation of VEGF and
angiogenesis by the transcriptional coactivator PGC-1alpha. Nature 2008;451:1008–1012. [PubMed:
18288196]
17. Gaillard S, Dwyer MA, McDonnell DP. Definition of the molecular basis for estrogen receptor-related
receptor-alpha-cofactor interactions. Mol Endocrinol 2007;21:62–76. [PubMed: 17053040]
18. Banks, E. The epidemiology of ovarian cancer. In: Bartlett, JMS., editor. Ovarian Cancer, Methods
and Protocols. Humana Press; Totowa, NJ: 2000. p. 3-6.
19. Bosetti C, Rossi M, McLaughlin JK, Negri E, Talamini R, et al. Flavonoids and the risk of renal cell
carcinoma. Cancer Epidemiol Biomarkers Prev 2007;16:98–101. [PubMed: 17220336]
20. Gonzalez CA, Riboli E. Diet and cancer prevention: where we are, where we are going. Nutr Cancer
2006;56:225–231. [PubMed: 17474869]
21. Holick CN, Giovannucci EL, Rosner B, Stampfer MJ, Michaud DS. Prospective study of intake of
fruit, vegetables, and carotenoids and the risk of adult glioma. Am J Clin Nutr 2007;85:877–886.
[PubMed: 17344512]
22. Mink PJ, Scrafford CG, Barraj LM, Harnack L, Hong CP, et al. Flavonoid intake and cardiovascular
disease mortality: a prospective study in postmenopausal women. Am J Clin Nutr 2007;85:895–909.
[PubMed: 17344514]
23. Bosetti C, Bravi F, Talamini R, Parpinel M, Gnagnarella P, et al. Flavonoids and prostate cancer risk:
a study in Italy. Nutr Cancer 2006;56:123–127. [PubMed: 17474856]
24. Theodoratou E, Kyle J, Cetnarskyj R, Farrington SM, Tenesa A, et al. Dietary flavonoids and the risk
of colorectal cancer. Cancer Epidemiol Biomarkers Prev 2007;16:684–693. [PubMed: 17416758]
25. Adhami VM, Malik A, Zaman N, Sarfaraz S, Siddiqui IA, et al. Combined inhibitory effects of green
tea polyphenols and selective cyclooxygenase-2 inhibitors on the growth of human prostate cancer
cells both in vitro and in vivo. Clin Cancer Res 2007;13:1611–1619. [PubMed: 17332308]
26. Plochmann K, Korte G, Koutsilieri E, Richling E, Riederer P, et al. Structure-activity relationships
of flavonoid-induced cytotoxicity on human leukemia cells. Arch Biochem Biophys 2007;460:1–9.
[PubMed: 17353006]
27. Choi EJ, Kim T, Lee MS. Pro-apoptotic effect and cytotoxicity of genistein and genistin in human
ovarian cancer SK-OV-3 cells. Life Sci 2007;80:1403–1408. [PubMed: 17291540]
28. Hung H. Inhibition of estrogen receptor alpha expression and function in MCF-7 cells by kaempferol.
J Cell Physiol 2004;198:197–208. [PubMed: 14603522]
29. Leung HW, Lin CJ, Hour MJ, Yang WH, Wang MY, et al. Kaempferol induces apoptosis in human
lung non-small carcinoma cells accompanied by an induction of antioxidant enzymes. Food Chem
Toxicol 2007;45:2005–2013. [PubMed: 17583406]
30. Sharma V, Joseph C, Ghosh S, Agarwal A, Mishra MK, et al. Kaempferol induces apoptosis in
glioblastoma cells through oxidative stress. Mol Cancer Ther 2007;6:2544–2453. [PubMed:
17876051]
31. Cortes JR, Perez-G M, Rivas MD, Zamorano J. Kaempferol inhibits IL-4-induced STAT6 activation
by specifically targeting JAK3. J Immunol 2007;179:3881–3887. [PubMed: 17785825]
32. García-Mediavilla V, Crespo I, Collado PS, Esteller A, Sánchez-Campos S, et al. The anti-
inflammatory flavones quercetin and kaempferol cause inhibition of inducible nitric oxide synthase,
cyclooxygenase-2 and reactive C-protein, and down-regulation of the nuclear factor kappaB pathway
in Chang liver cells. Eur J Pharmacol 2007;557:221–229. [PubMed: 17184768]
33. Gates MA, Tworoger SS, Hecht JL, De Vivo I, Rosner B, et al. A prospective study of dietary flavonoid
intake and incidence of epithelial ovarian cancer. Int J Cancer 2007;121:2225–2232. [PubMed:
17471564]
34. Kolfschoten GM, Hulscher TM, Duyndam MC, Pinedo HM, Boven E. Variation in the kinetics of
caspase-3 activation, Bcl-2 phosphorylation and apoptotic morphology in unselected human ovarian
Luo et al. Page 9
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
cancer cell lines as a response to docetaxel. Biochem Pharmacol 2002;63:733–743. [PubMed:
11992642]
35. Duyndam MC, van Berkel MP, Dorsman JC, Rockx DA, Pinedo HM, et al. Cisplatin and doxorubicin
repress vascular endothelial growth factor expression and differentially down-regulate hypoxia-
inducible factor I activity in human ovarian cancer cells. Biochem Pharmacol 2007;74:191–201.
[PubMed: 17498666]
36. Risch HA, Jain M, Marrett LD, Howe GR. Dietary fat intake and risk of epithelial ovarian cancer. J
Natl Cancer Inst 1994;86:1409–1415. [PubMed: 8072035]
37. Ferrara N. The role of VEGF in the regulation of physiological and pathological angiogenesis. EXS
2005;94:209–231. [PubMed: 15617481]
38. Coultas L, Chawengsaksophak K, Rossant J. Endothelial cells and VEGF in vascular development.
Nature 2005;438:937–945. [PubMed: 16355211]
39. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971;285:1182–1186.
[PubMed: 4938153]
40. Risau W. Mechanisms of angiogenesis. Nature 1997;386:671–674. [PubMed: 9109485]
41. Byrne AM, Bouchier-Hayes DJ, Harmey JH. Angiogenic and cell survival functions of vascular
endothelial growth factor (VEGF). J Cell Mol Med 2005;9:777–794. [PubMed: 16364190]
42. Nishida N, Yano H, Komai K, Nishida T, Kamura T, et al. Vascular endothelial growth factor C and
vascular endothelial growth factor receptor 2 are related closely to the prognosis of patients with
ovarian carcinoma. Cancer 2004;101:1364–1374. [PubMed: 15368324]
43. Jiang BH, Liu LZ. AKT signaling in regulating angiogenesis. Curr Cancer Drug Targets 2008;8:19–
26. [PubMed: 18288940]
44. Hoeben A, Landuyt B, Highley MS, Wildiers H, Van Oosterom AT, et al. Vascular Endothelial
Growth Factor and Angiogenesis. Pharmacol Rev 2004;56:549–580. [PubMed: 15602010]
45. Kallen J, Schlaeppi JM, Bitsch F, Filipuzzi I, Schilb A, et al. Evidence for ligand-independent
transcriptional activation of the human estrogen-related receptor alpha (ERRalpha): crystal structure
of ERRalpha ligand binding domain in complex with peroxisome proliferator-activated receptor
coactivator-1alpha. J Biol Chem 2004;279:49330–49337. [PubMed: 15337744]
Luo et al. Page 10
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 1.
Kaempferol’s effect on cell viability, VEGF expression, tumorigenesis, and angiogenesis in
ovarian cancer cell lines. A: Cells (8 × 103/well) were seeded in 96-well plates, incubated for
16 h, and treated with kaempferol for 24 h. Cell viability was colorimetrically determined by
a MTS-based method and expressed as percentages of control. Data represents mean ± SE from
3 independent experiments. B: Cells (5 × 105) were seeded in 60-mm dishes, incubated 16 h,
and treated with 40-μM kaempferol for 0 to 24 h. Cells were harvested in TRIzol Reagent and
stored in 80 °C until analysis. RNA was extracted; reverse transcribed with AMV reverse
transcriptase; and quantitated by SYBR Green-based, real-time PCR. VEGF mRNA levels
were normalized by GAPDH abundance and expressed as percentages of control. Data
Luo et al. Page 11
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
represents mean ± SE from 2 independent experiments. C: Cells (5 × 105) were seeded in 60-
mm dishes, incubated 16 h, and treated with 0 to 40 μM kaempferol for 24 h. RNA was extracted
with TRIzol Reagent, reverse-transcribed with AMV reverse transcriptase, and quantitated by
SYBR Green-based, real-time PCR. VEGF mRNA levels were normalized by GAPDH
abundance and expressed as percentages of control. Data represents mean ± SE from 3
independent experiments. D: Cells (8 × 103/well) were seeded in 96-well plates, incubated for
16 h, and treated with kaempferol for 24 h. Culture supernates were collected and analyzed for
VEGF165 by ELISA, and cell numbers were determined by MTS-based assay. VEGF levels
were normalized by cell numbers and expressed as percentages of control. Data represents
mean ± SE from 4 independent experiments. E: OVCAR-3 cells (3 × 106) were suspended in
100 μ l serum-free medium at 4°C, mixed with 50 μl cold BD Matrigel, supplemented with or
without 20-μM kaempferol at 4°C, implanted in the chorioallantoic membrane of 9-day-old
chicken embryos, and incubated for 5 days at 37.5°C before tumors were weighed and counted
for blood vessels. Data represents mean ± SE from 5 chicken embryos. *P < 0.05 as compared
to control. **P < 0.01 as compared to control.
Luo et al. Page 12
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 2.
Kaempferol’s effect on HIF gene expression. A and B: Cells (5 × 105) were seeded in 60-mm
dishes, incubated 16 h, and treated with kaempferol for 24 h. RNA was extracted with TRIzol
Reagent, reverse-transcribed with AMV reverse transcriptase, and quantitated by SYBR
Green-based, real-time PCR. HIF-1α and HIF-1β mRNA levels were normalized by GAPDH
abundance and expressed as percentages of control. Data represents mean ± SE from 3
independent experiments. C and D: Cells (5 × 105) were seeded in 60-mm dishes, incubated
for 16 h, and treated with kaempferol for 24 h. Cells were harvested with RIPA buffer, and cell
lysates were separated by SDS-PAGE and blotted into nitrocellulose membrane for
immunodetection. Chemiluminescent signals were captured by x-ray film and quantitated by
imaging software. HIF-1α and HIF-1β protein levels were normalized by GAPDH and
expressed as percentages of control. Typical blots were shown for both cell lines, and data
represents mean ± SE from 3 independent experiments. *P < 0.05 as compared to control.
**P < 0.01 as compared to control.
Luo et al. Page 13
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 3.
Kaempferol’s effect on p-AKT and p-ERK levels. A and B: Cells (5 × 105) were seeded in 60-
mm dishes, incubated for 16 h, and treated with kaempferol for 24 h. Cell lysates were analyzed
for p-AKT and p-ERK with ELISA, and for total protein with BCA assay. Levels of p-AKT
and p-ERK were normalized by total protein. Data represents mean ± SE from 4 independent
experiments. *P < 0.05 as compared to control.
Luo et al. Page 14
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 4.
Kaempferol’s effect on PPARGC1A and ESRRA mRNA level. A and B: Cells (5 × 105) were
seeded in 60-mm dishes, incubated 16 h, and treated with kaempferol for 24 h. RNA was
extracted with TRIzol Reagent, reverse-transcribed with AMV reverse transcriptase, and
quantitated by SYBR Green-based, real-time PCR. PPARGC1A and ESRRA mRNA levels
were normalized by GAPDH abundance and expressed as percentages of control. Data
represents mean ± SE from 3 independent experiments. **P < 0.01 as compared to control.
Luo et al. Page 15
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 5.
Proposed mechanism for kaempferol’s inhibition of angiogenesis and proliferation in ovarian
cancer cells. Connector lines represent established pathways. Arrows indicate regulations by
kaempferol treatment in experimental results. PPARGC1A, peroxisome proliferator-activated
receptor gamma coactivator 1 alpha; ESRRA, estrogen-related receptor alpha; HIF-1α,
hypoxia-inducible factor 1 alpha; VEGF, vascular endothelial growth factor.
Luo et al. Page 16
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Luo et al. Page 17
TABLE 1
Primer sequences of genes analyzed by qRT-PCRa
Gene GenBank Accession Primers Amplicon Size
VEGF NM 001025368 AAG GAG GAG GGC AGA ATC AT 226
ATC TGC ATG GTG ATG TTG GA
GAPDH NM 002046 CAT GAG AAG TAT GAC AAC AGC CT 113
AGT CCT TCC ACG ATA CCA AAG T
HIF-1αNM 001530 ATC CAT GTG ACC ATG AGG AAA TG 126
CTC GGC TAG TTA GGG TAC ACT T
HIF-1βNM 001668 CTG CCA ACC CCG AAA TGA CAT 117
GCC GCT TAA TAG CCC TCT GG
ESRRA NM 004451 GTC CAA AGG GTT CCT CGG AG 221
GGA TGC CAC ACC ATA GTG GTA
PPARGC1A NM 013261 TGG GTA GCC CAT CAA AAT GT 176
TGG TAC TTA CCA CGG CAT GA
aAbbreviations are as follows: qRT-PCR, quantitative reverse-transcription polymerase chain reaction; VEGF, vascular endothelial growth factor; GAPDH, glyderaldehyde-3-phosphate dehydrogenase;
HIF, hypoxia-inducible factor; ESRRA, estrogen-related receptor alpha; PPARGC1A, peroxisome proliferator-activated receptor gamma coactivator 1 alpha.
Nutr Cancer. Author manuscript; available in PMC 2009 October 30.
... The findings were consistent, indicating a strong cytotoxicity of KMP. Another focus of studies on KMP was ovarian cancer [34,37,40,41,57,60,79]. The results demonstrated positive effects, with KMP reducing cell viability and angiogenesis and increasing apoptosis, with cell cycle arrest at G2/M. ...
... The results demonstrated positive effects, with KMP reducing cell viability and angiogenesis and increasing apoptosis, with cell cycle arrest at G2/M. Luo et al. [34] suggested that KMP inhibits tumor angiogenesis in human ovarian cancer by suppressing vascular endothelial growth factor (VEGF) expression through a hypoxia-inducible factor (HIF)-dependent (AKT/HIF) pathway in ovarian cancer cells. In cervical cancer, KMP has been shown to reduce cell viability and increase apoptosis [55]. ...
... The main mediator of angiogenesis is VEGF. Evaluating the effect of KMP on ovarian cancer cell lines, Luo et al. [34] observed a significant reduction in VEGF and HIF-1, which are critical for adaptive vascular responses to ischemia/hypoxia. By targeting these key mediators, KMP may disrupt the angiogenic process, thereby limiting the blood supply to the tumor and impeding its growth. ...
Article
Full-text available
Simple Summary Kaempferol, a natural compound commonly found in fruits, vegetables, and plants, has gained interest within the scientific community because of its anticancer properties against different types of tumors. The results of this review reveal that kaempferol exerts anticancer effects on many types of tumor cells by different mechanisms, providing evidence of its potential as a cancer drug. Abstract Given the heterogeneity of different malignant processes, planning cancer treatment is challenging. According to recent studies, natural products are likely to be effective in cancer prevention and treatment. Among bioactive flavonoids found in fruits and vegetables, kaempferol (KMP) is known for its anti-inflammatory, antioxidant, and anticancer properties. This systematic review aims to highlight the potential therapeutic effects of KMP on different types of solid malignant tumors. This review was conducted following the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. Searches were performed in EMBASE, Medline/PubMed, Cochrane Collaboration Library, Science Direct, Scopus, and Google Scholar. After the application of study criteria, 64 studies were included. In vitro experiments demonstrated that KMP exerts antitumor effects by controlling tumor cell cycle progression, proliferation, apoptosis, migration, and invasion, as well as by inhibiting angiogenesis. KMP was also able to inhibit important markers that regulate epithelial–mesenchymal transition and enhanced the sensitivity of cancer cells to traditional drugs used in chemotherapy, including cisplatin and 5-fluorouracil. This flavonoid is a promising therapeutic compound and its combination with current anticancer agents, including targeted drugs, may potentially produce more effective and predictable results.
... Kaempferol, a plant-derived flavonoid widely distributed in natural food sources, including fruits and vegetables, has been reported to display a wide range of pharmacological activities, including anti-inflammatory properties [10][11][12][13]. It has been found to inhibit cancer cell proliferation and VEGF expression, induce caspase 3 activity in oral cavity cancer cells [14][15][16] and up-regulate pro-apoptotic bax and bad genes while downregulating anti-apoptotic genes bcl-2 and bcl-Xl in lung cancer cells [17,18]. Evidence indicates that kaempferol reverses the drug resistance to 5-Fluorouracil in HCT8-R cells by promoting the expression of microRNA-326, leading to the inhibition of glycolysis pathway by targeting pyruvate kinase M2 (PKM2) [19]. ...
Article
Full-text available
Colorectal cancer (CRC) is the third most diagnosed and highly fatal malignancy, presenting serious health concerns worldwide. The search for an effective cure for CRC is challenging and poses a serious concern. Kaempferol is a potent anti-cancerous bioactive compound often suggested for treating various cancers, including CRC. However, its underlying molecular mechanism against CRC remains unclear. The present study delves into kaempferol's molecular pathways and underlying molecular mechanisms against CRC targets. The target protein-coding genes for kaempferol were retrieved, and the CRC-associated genes were curated. Twelve common targets with a disease specificity index of > 0.6 were validated for their protein expression at different stages of CRC. Over-expressed USP1, SETD7, POLH, TDP1 and RACGAP1 were selected for further studies. The binding affinities of kaempferol to the corresponding proteins were evaluated using molecular docking and Molecular Dynamics (MD) simulations. SETD7 exhibited the highest binding affinity with the lowest binding energy (− 8.06 kcal/mol). Additionally, the MD simulation, and MM-PBSA conferred SETD7-kaempferol complex had the least root-mean-square deviation with lower interaction energy and higher conformational stability. The protein–protein interaction of SETD7 constructed revealed direct interactors, namely, DNMT1, FOXO1, FOXO3, FOXO4, H3-3B, H3-4, H3C12, H3C13, SETD7, SIRT1 and TP53, have a potential role in cancer progression through FOXO signalling. In summary, our study revealed kaempferol's multi-target and synergistic effect on multiple CRC targets and its underlying mechanisms. Finally, the study recommends in-vitro and in-vivo trials for validation of anti-cancerous drugs for CRC.
... However, β-carboline-benzoquinolizidine alkaloid deoxytubulosine [16] and kaempferol [17] are two of the significant components found in Alangium salvifolium, which is why it was selected for this study over other plant species. Kaempferol, which is present in leaves, is responsible for its anticancer effect whereas deoxytubulosine exhibits DNA-damaging activity and dihydrofolate reductase inhibition [18,19]. Although nearly every part of this plant species is considered restorative, particularly the leaves and flowers, has been established to have therapeutic values. ...
Article
Full-text available
Green synthesis of silver/silver nanoparticles from the combination of Pandanus foetidus and Alangium salvifolium was confirmed by an absorption peak at 455 nm. Fourier transform infrared spectroscopy analysis showed the existence of multiple possible functional groups through C-O, N-H and O-H stretching. The presence of several elements along with silver has been found by Energy-dispersive X-ray spectroscopy, while a high-intensity reflection peak at 38.45◦ revealed information regarding the crystalline nature of synthesized nanoparticles. Scanning electron and Transmission electron microscopy determined their spherical size to be 23.5 nm. With moderate toxicity (LC50 value of 282.74 μg/ml), AgNPs inhibited 77.7 % and 58.4 % of Ehrlich ascites carcinoma (EAC) cell growth in Swiss albino mice at dose levels of 1 mg/kg/day and 2 mg/kg/day, respectively and concurrently brought back the hematological parameters (RBC, WBC, SGOT, SGPT and hemoglobin) to normal levels. The phytochemical potency of synthesized nanoparticles was revealed with an IC50 value of 33 μg/ml, compared to the corresponding combined plant extract, with a total phenol and flavonoid content of 47.03 ± 1.52 mg/g of gallic acid and 404.13 ± 1.22 mg/g of catechin, respectively. The AgNPs demonstrated analgesic activity (85.8 and 77.49 %) with anti-inflammatory action (85.58 and 78.95 %) at doses of 100 and 50 mg/kg body weight, corresponding to their standards. AgNPs possessed a substantial inhibitory effect of 51.01 % on Shigella boydii (ATCC 231903) at a dose of 50 μg/ml while Staphylococcus aureus (ATCC 25923), Bacillus cereus (ATCC 14579), and Escherichia coli (ATCC 27853) were less susceptible.
... It downregulates the phosphorylated ERK, and the levels of cMyc and NF-κB while promoting p21 expression [94]. Another study also reported that kaempferol's anti-angiogenic effect and inhibition of VEGF level is performed by reducing HIF-1α expression via two different pathways, Akt/HIF and ESRRA, in which ESRRA, an orphan nuclear receptor, shares significant sequence similarity and engages in intense crosstalk with estrogen receptors [95]. Additionally, kaempferol was found to significantly suppress both VEGF and FGF signaling pathways, leading to a direct reduction in VEGFR2 expression [96]. ...
Article
Full-text available
The formation of new blood vessels, known as angiogenesis, significantly impacts the development of multiple types of cancer. Consequently, researchers have focused on targeting this process to prevent and treat numerous disorders. However, most existing anti-angiogenic treatments rely on synthetic compounds and humanized monoclonal antibodies, often expensive or toxic, restricting patient access to these therapies. Hence, the pursuit of discovering new, affordable, less toxic, and efficient anti-angiogenic compounds is imperative. Numerous studies propose that natural plant-derived products exhibit these sought-after characteristics. The objective of this review is to delve into the anti-angiogenic properties exhibited by naturally derived flavonoids from plants, along with their underlying molecular mechanisms of action. Additionally, we summarize the structure, classification, and the relationship between flavonoids with their signaling pathways in plants as anti-angiogenic agents, including main HIF-1α/VEGF/VEGFR2/PI3K/AKT, Wnt/β-catenin, JNK1/STAT3, and MAPK/AP-1 pathways. Nonetheless, further research and innovative approaches are required to enhance their bioavailability for clinical application.
... Quercetin, on the other hand, not only has its inherent cytotoxic property but it also can potentiate the cytotoxicity of luteolin [54,56]. Similarly, kaempferol have also demonstrated both cytotoxic and antiangiogenic [57,58]. However, no concrete findings directly confirm the cytotoxic and antiangiogenic properties of caffeic anhydride and methyl rose marinate. ...
Article
Full-text available
In recent years, there has been a significant demand for natural products as a mean of disease prevention or as an alternative to conventional medications. The driving force for this change is the growing recognition of the abundant presence of valuable bioactive compounds in natural products. On recent years Actinia arguta fruit, also known as kiwiberry, has attracted a lot of attention from scientific community due to its richness in bioactive compounds, including phenolic compounds, organic acids, vitamins, carotenoids and fiber. These bioactive compounds contribute to the fruit’s diverse outstanding biological activities such as antioxidant, anti-inflammatory, neuroprotective, immunomodulatory, and anti-cancer properties. Due to these properties, the fruit may have the potential to be used in the treatment/prevention of various types of cancer, including glioblastoma. Glioblastoma is the most aggressive form of brain cancer, displaying 90 % of recurrence rate within a span of 2 years. Despite the employment of an aggressive approach, the prognosis remains unfavorable, emphasizing the urgent requirement for the development of new effective treatments. The preclinical evidence suggests that kiwiberry has potential impact on glioblastoma by reducing the cancer self-renewal, modulating the signaling pathways involved in the regulation of the cell phenotype and metabolism, and influencing the consolidation of the tumor microenvironment. Even though, challenges such as the imprecise composition and concentration of bioactive compounds, and its low bioavailability after oral administration may be drawbacks to the development of kiwiberry-based treatments, being urgent to ensure the safety and efficacy of kiwiberry for the prevention and treatment of glioblastoma. This review aims to highlight the potential impact of A. arguta bioactive compounds on glioblastoma, providing novel insights into their applicability as complementary or alternative therapies.
Article
After the developing embryo has formed a primary vascular plexus by a process termed vasculogenesis, further blood vessels are generated by both sprouting and non-sprouting angiogenesis, which are progressively pruned and remodelled into a functional adult circulatory system. Recent results, particularly from the study of mice lacking some of the signalling systems involved, have greatly improved our understanding of the molecular basis underlying these events, and may suggest new approaches for treating conditions such as cancer that depend on angiogenesis.
Article
BACKGROUND The vascular endothelial growth factor (VEGF) family and VEGF receptors (VEGFR) play an essential role in the angiogenesis of both pathologic and nonpathologic conditions. However, the prognostic significance of VEGF and VEGFR expression in ovarian carcinoma is unclear.METHODS The tissue expression levels of VEGF-A, VEGF-C, VEGFR-2, and VEGFR-3 in 80 specimens of ovarian carcinoma were examined immnohistochemically. The results obtained were analyzed clinicopathologically.RESULTSVEGF-A, VEGF-C, VEGFR-2, and VEGFR-3 were expressed both in tumor cells and in adjacent endothelial cells of blood and lymph vessels. The tissue expressions of VEGF-C and VEGFR-2 were correlated significantly with tumor extension, including peritoneal metastases outside the pelvic cavity (P = 0.0010 and P = 0.0008, respectively), lymph node metastases (P = 0.0030 and P = 0.0018, respectively), and positive ascitic cytology (P = 0.025 and P = 0.0016, respectively). Conversely, there was no significant correlation between VEGF-A and VEGFR-3 expression and clinicopathologic features of ovarian carcinoma. Logistic regression analysis revealed that the expressions of VEGF-C and VEGFR-2 also were independent risk factors for peritoneal and lymph node metastases. Survival curves determined by the Kaplan–Meier method and in univariate analysis demonstrated that high expression levels of VEGF-C and VEGFR-2 were associated with the 5-year survival rate. In multivariate analysis, high expression levels of VEGF-C and VEGFR-2 emerged as independent indicators for disease-specific survival.CONCLUSIONS High tissue expression of VEGF-C and VEGFR-2 reflects the aggressiveness of the spread of tumor in ovarian carcinoma. Thus, both have predictive value for identifying high-risk patients who have a poor prognosis. Cancer 2004. © 2004 American Cancer Society.
Article
Ovarian cancer is the most common fatal cancer of the female reproductive tract in industrialized countries. At the time of writing, it is the fourth most common cause of cancer death in women in the U.K., after breast, lung, and colorectal cancer, with a lifetime risk of approximately 2% (1). It tends to present at an advanced stage, with limited prospects for treatment and generally poor survival.
Article
This article has no abstract; the first 100 words appear below. THE growth of solid neoplasms is always accompanied by neovascularization. This new capillary growth is even more vigorous and continuous than a similar outgrowth of capillary sprouts observed in fresh wounds or in inflammation.¹ Many workers have described the association between growing solid malignant tumors and new vessel growth.²³⁴⁵⁶ However, it has not been appreciated until the past few years that the population of tumor cells and the population of capillary endothelial cells within a neoplasm may constitute a highly integrated ecosystem. In this ecosystem the mitotic index of the two cell populations may depend upon each other. Tumor cells . . . Supported by a grant (5 RO1 CA08185–06) from the National Cancer Institute, a grant from the American Cancer Society, National Chapter (IC-28), and gifts from the Merck Company and the Alza Corporation. Source Information From the Department of Surgery, Children's Hospital Medical Center and Harvard Medical School, Boston, Massachusetts 02115.
Article
This editorial argues the cause-and-effect relationship between in utero exposure to diethylstilbestrol (DES) and incidence of vaginal adenocarcinoma among young women. It is conjectured that the mechanism by which DES induces tumors is a result of transplacental carcinogenesis; i.e., DES causes a malignant change in any fetal cell of future vaginal tissue, resulting in a genetic defect which may not be realized until puberty, when endogenous hormone production acts as a promoter of the malignancy initiated by DES exposure during gestation. If this theory is correct, local progestational therapy may arrest future adenocarcinoma cases. Though it is obvious that DES therapy must be avoided in the future in the population of pregnant women, of more concern is the presence of residual DES in foodstuffs, particularly livestock meat. A ban of such diet supplementation of human foodstuffs is called for.
Article
Chemotherapy combinations that include an alkylating agent and a platinum coordination complex have high response rates in women with advanced ovarian cancer. Such combinations provide long-term control of disease in few patients, however. We compared two combinations, cisplatin and cyclophosphamide and cisplatin and paclitaxel, in women with ovarian cancer. We randomly assigned 410 women with advanced ovarian cancer and residual masses larger than 1 cm after initial surgery to receive cisplatin (75 mg per square meter of body-surface area) with either cyclophosphamide (750 mg per square meter) or paclitaxel (135 mg per square meter over 24 hours). Three hundred eighty-six women met all the eligibility criteria. Known prognostic factors were similar in the two treatment groups. Alopecia, neutropenia, fever, and allergic reactions were reported more frequently in the cisplatin-paclitaxel group. Among 216 women with measurable disease, 73 percent in the cisplatin-paclitaxel group responded to therapy, as compared with 60 percent in the cisplatin-cyclophosphamide group (P = 0.01). The frequency of surgically verified complete response was similar in the two groups. Progression-free survival was significantly longer (P < 0.001) in the cisplatin-paclitaxel group than in the cisplatin-cyclophosphamide group (median, 18 vs. 13 months). Survival was also significantly longer (P < 0.001) in the cisplatin-paclitaxel group (median, 38 vs. 24 months). Incorporating paclitaxel into first-line therapy improves the duration of progression-free survival and of overall survival in women with incompletely resected stage III and stage IV ovarian cancer.
Article
Evidence exists that dietary fat may be a contributory factor in the development of hormone-related cancers such as ovarian cancer. Previous studies have demonstrated significantly higher circulating-estrogen levels among nonvegetarian women than among vegetarian women; the increase correlated directly with consumption of saturated fat. However, the contribution that dietary fat plays in the development of hormone-related cancers remains unresolved. Our purpose was to evaluate whether saturated fat intake increases the risk of ovarian cancer development. Population-based sampling was used to acquire cases and controls over a 3-year period from the study area, which included the highly populated region surrounding the western end of Lake Ontario, Canada. From the Ontario Cancer Registry, all histologically confirmed, primary malignant or borderline malignant epithelial ovarian tumors first diagnosed from November 1989 through October 1992 among study-area residents aged 35-79 years were determined. In total, 631 eligible case subjects were identified, of whom 450 (71.3%) were interviewed concerning reproduction and diet; 564 randomly selected population control subjects were similarly interviewed. From the quantitative diet-history information, average daily intakes of macronutrients and micronutrients were calculated. Unconditional continuous logistic regression methods were used for analysis, with adjustment for age at interview, number of full-term pregnancies, years of oral contraceptive use, and total daily caloric intake. Saturated fat consumption was associated with increasing risk of ovarian cancer (odds ratio [OR] = 1.20 for each 10 g/day of intake; 95% confidence interval [CI] = 1.03-1.40; one-sided P = .0082). No relationship was seen with intake of unsaturated fats. Egg consumption also appeared related to increased risk (OR = 1.42 for each 100 mg of egg cholesterol per day; 95% CI = 1.18-1.72; two-sided P = .0002), though this association may have resulted from disease-related changes in the dietary practices of case subjects prior to diagnosis. Consumption of vegetable fiber (but not fruit or cereal fiber) was associated with decreased risk (OR = 0.63 for each 10 g/day; 95% CI = 0.49-0.80; two-sided P = .0001). All three nutrients (saturated fat, egg cholesterol, and vegetable fiber) remained statistically significant when included in the same regression model. Diet may contribute to risk of ovarian cancer development. If confirmed in further studies, this association may allow women to appreciably lower their risk of ovarian cancer through dietary modifications: reducing the intake of saturated fats and eating more vegetables.
Article
Ovarian germ cell malignancies are neoplasms derived from primitive germ cells of the embryonic gonad. These tumors are highly malignant, rapidly growing, and typically occur in young women. The prognosis for patients with ovarian non-dysgerminomatous germ cell malignancies was bleak before the introduction of modern combination chemotherapy. The evolution of modern chemotherapy transformed these virulent malignancies into highly curable ones. In the early 1970s, the combination of vincristine, actinomycin D, and cyclophosphamide (VAC) emerged as the first effective therapy. The efficacy of cisplatin, vinblastine, and bleomycin (PVB) was documented in treatment of men with testicular cancer and subsequently became standard treatment for women with ovarian germ cell malignancies. Bleomycin, etoposide, and cisplatin (BEP) are shown to have equal efficacy and less toxicity in the treatment of ovarian germ cell malignancies. Experience at Yale University suggested that patients with ovarian germ cell malignancies could be managed by using tumor histology to determine the type of chemotherapy, and determining treatment duration by serial assays of circulating tumor markers or by International Federation of Gynecologists and Obstetricians' staging. Preservation of reproductive function is appropriate for all patients with early stage disease and selected patients with more advanced disease.