ArticlePDF Available

Concentration of endogenous estrogens and estrogen metabolites in the NCI-60 human tumor cell lines

Authors:
  • Maranatha Baptist University

Abstract and Figures

Endogenous estrogens and estrogen metabolites play an important role in the pathogenesis and development of human breast, endometrial, and ovarian cancers. Increasing evidence also supports their involvement in the development of certain lung, colon and prostate cancers. In this study we systemically surveyed endogenous estrogen and estrogen metabolite levels in each of the NCI-60 human tumor cell lines, which include human breast, central nerve system, colon, ovarian, prostate, kidney and non-small cell lung cancers, as well as melanomas and leukemia. The absolute abundances of these metabolites were measured using a liquid chromatography-tandem mass spectrometry method that has been previously utilized for biological fluids such as serum and urine. Endogenous estrogens and estrogen metabolites were found in all NCI-60 human tumor cell lines and some were substantially elevated and exceeded the levels found in well known estrogen-dependent and estrogen receptor-positive tumor cells such as MCF-7 and T-47D. While estrogens were expected to be present at high levels in cell lines representing the female reproductive system (that is, breast and ovarian), other cell lines, such as leukemia and colon, also contained very high levels of these steroid hormones. The leukemia cell line RMPI-8226 contained the highest levels of estrone (182.06 pg/106 cells) and 17β-estradiol (753.45 pg/106 cells). In comparison, the ovarian cancer cell line with the highest levels of these estrogens contained only 19.79 and 139.32 pg/106 cells of estrone and 17β-estradiol, respectively. The highest levels of estrone and 17β-estradiol in breast cancer cell lines were only 8.45 and 87.37 pg/106 cells in BT-549 and T-47D cells, respectively. The data provided evidence for the presence of significant amounts of endogenous estrogens and estrogen metabolites in cell lines not commonly associated with these steroid hormones. This broad discovery of endogenous estrogens and estrogen metabolites in these cell lines suggest that several human tumors may be beneficially treated using endocrine therapy aimed at estrogen biosynthesis and estrogen-related signaling pathways.
Content may be subject to copyright.
RESEARCH Open Access
Concentration of endogenous estrogens and
estrogen metabolites in the NCI-60 human tumor
cell lines
Xia Xu and Timothy D Veenstra
*
Abstract
Background: Endogenous estrogens and estrogen metabolites play an important role in the pathogenesis and
development of human breast, endometrial, and ovarian cancers. Increasing evidence also supports their
involvement in the development of certain lung, colon and prostate cancers.
Methods: In this study we systemically surveyed endogenous estrogen and estrogen metabolite levels in each of
the NCI-60 human tumo r cell lines, which include human breast, central nerve system, colon, ovarian, prostate,
kidney and non-small cell lung cancers, as well as melanomas and leukemia. The absolute abundances of these
metabolites were measured using a liquid chromatography-tandem mass spectrometry method that has been
previously utilized for biological fluids such as serum and urine.
Results: Endogenous estrogens and estrogen metabolites were found in all NCI-60 human tumor cell lines and
some were substantially elevated and exceeded the levels found in well known estrogen-dependent and estrogen
receptor-positive tumor cells such as MCF-7 and T-47D . While estrogens were expected to be present at high
levels in cell lines representing the female reproductive system (that is, breast and ovarian), other cell lines, such as
leukemia and colon, also contained very high levels of these steroid hormones. The leuke mia cell line RMPI-8226
contained the highest levels of estrone (182.06 pg/10
6
cells) and 17b-estradiol (753.45 pg/10
6
cells). In comparison,
the ovarian cancer cell line with the highest levels of these estrogens contained only 19.79 and 139.32 pg/10
6
cells
of estrone and 17b-estradiol, respectively. The highest levels of estrone and 17b-estradiol in breast cancer cell lines
were only 8.45 and 87.3 7 pg/10
6
cells in BT-549 and T-47D cells, respectively.
Conclusions: The data provided evidence for the pres ence of significant amounts of endogenous estrogens and
estrogen metabolites in cell lines not commonly associated with these steroid hormones. This broad discovery of
endogenous estrogens and estrogen metabolites in these cell lines suggest that several human tumors may be
beneficially treated using endocrine therapy aimed at estrogen biosynthesis and estrogen-related signaling
pathways.
Background
Endogenous estrogens and estrogen metabolites (EMs)
have long been associated with carcinogenesis and devel-
opment of several hormone-dependent human carcino-
mas, such as breast, endometrial, and ovarian cancers
[1,2]. Increasing evidence suggests that these metabolites
may be involved in the pathogenesis and development of
human lung [3,4] and colon [5] cancers as well as prostate
cancer [6]. Historically, the major primary function of 17b
estradiol (E
2
) was the development of female secondary
sexual characteristics and regulation of reproductive func-
tion. Today it is recognized that E
2
exerts some effect on
almost every organ in the body [7]. The effects of E
2
and
other estrogens have expanded to include roles in neurolo-
gical function [8], retinal degenerative disease [9], cardio-
vascular health [10], and even sleep regulation [11].
Given the well documented mitogenic and possible gen-
otoxic nature of endogenous estrogens and EMs [2,12,13],
potential involvement of EMs in the carcinogenesis of
an eve n greater va riety of human tumors is conceivable.
* Correspondence: veenstrat@mail.nih.go v
Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc.,
National Cancer Institute at Frederick, Frederick, MD 21702, USA
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
© 2012 Xu and Veenstra; licensee BioMed Central Ltd . This is an open access article distributed under the terms of the Creative
Commons Attribution License (http://cr eativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
reproduction in any medium, provi ded the original work is prope rly cit ed.
For example, some studies have suggested that estrogen
may be involved in the development of skin cancer as skin
keratinocytes possess estrogen receptors (ERs) [14], and
oral contraceptives and hormone therapy decrease acne
[15] and skin aging [16], respectively. Epidemiological stu-
dies examining assoc iations between hormone therapy
and melanoma risk have not been entirely conclusive,
although some studies hav e shown a link between hor-
mone use and increased risk of melanoma [17,18]. Epide-
miologically, estrogens have also been linked to colon
cancer, as men are more likely to develop this disease and
hormon e replacement therapy has been shown to reduce
the risk of this cancer in women [19].
Owing to th e pervasive nature of the functions of EMs,
their effect on cancers may be more pronounced than pre-
viously thought. Determining EM roles in various cancers
requires a lot of information, including tumor receptor
status, aromatase activities, and the levels of these com-
pounds within cells. Towards this aim, we systemically
surveyed human tumor cell EM levels by using NCI-60
human tumor cell lines, including human b reast, central
nervous system (CNS), colon, ovarian, prostate, kidney,
melanoma, leukemia, and non-small cell lung cancers.
Detailed EM profiles in NCI-60 human tumor cell lines
are summarized and reported in this manuscript.
Materials and methods
Reagents and materials
Cell pellets from NCI-60 human tumor cell lines were
obtained from Developmental Therapeutics Program,
NCI/NIH. Fifteen estrogens and EMs, including estro ne
(E
1
), estradiol (E
2
), estriol (E
3
), 16-epiestriol (16-epiE
3
),
17-epiestriol (17-epiE
3
), 16-ketoestradiol (16-ketoE
2
),
16a-hydroxyestrone (16a-OHE
1
), 2-methoxyestrone
(2-MeOE
1
), 4-methoxyestrone (4-MeOE
1
), 2-hydroxyes-
trone-3-methyl ether (3-MeOE
1
), 2-methoxyestradiol
(2-MeOE
2
), 4-methoxyestradiol (4-MeOE
2
), 2-hydro-
xyestrone (2-OHE
1
), 4-hydr oxyestrone (4-OHE
1
), and
2-hydroxyestradiol (2-OHE
2
)wereobtainedfromStera-
loids, Inc. (Newport, RI, USA). Stable isotope-labeled
estrogens (SI-EM), including estradiol-13,14,15,16,17,18-
13
C
6
(
13
C
6
-E
2
) and estrone-13,14,15,16,17,18-
13
C
6
(
13
C
6
-E
1
) were purchased from Cambridge Isotope
Laboratories, Inc. (Andover, MA, USA); estriol-2,4,17-d
3
(d
3
-E
3
), 2-hydroxyestradiol-1,4,16,16,17 -d
5
(d
5
-2-OHE
2
),
and 2-methoxyestradiol-1,4,16,16,17-d
5
(d
5
-2-MeOE
2
),
were obtained from C/D/N Isotopes, Inc. (Pointe-Claire,
Quebec, Canada). 16-Epiestriol-2,4,16-d
3
(d
3
-16-epiE
3
)
was purchased from Medical Isotopes, Inc. (Pelham,
NH, USA). All EM a nd SI-EM analytical standards have
reported chemical and isotopic purity 98%, and were
used without further purification. Dichloromethane,
methanol and formic acid were obtained from EM
Science (Gibbstown, NJ, USA). Glacial acetic acid,
sodium bicarbonate, and L-ascorbic acid were purchased
from JT Baker (Phillipsburg, NJ, USA) and sodium
hydroxide and sodium acetate were purchased from
Fisher Scientific (Fair Lawn, NJ, USA). Dansyl chloride
and acetone were purchased from Aldrich Chemical Co.
(Milwaukee, WI, USA). All chemicals and solvents used
in this study were HPLC or reagent grade unless other-
wise noted.
Preparation of stock and working standard solutions and
calibration standards
Stock solutions o f EMs and SI-EMs were each pr epared
at 80 μg/ml by dissolving 2 mg of the estrogen powders
in met hanol with 0.1% L-ascorbic acid to a final volume
of 25 ml in a volumetric flask. The stock solutions are
stable for at least two months while stored at - 20°C.
Stock solutions were analyzed at the beginning of each
analysis to verify no time-dependent degradation of the
EM and SI-EM standards had occurred. Working stan-
dards of EMs and SI-EMs at 8 ng/ml were prepared b y
dilutions of the stock solutions using methanol wit h
0.1% L-ascorbic acid.
MCF-10A cell lysate with no detectable levels of EMs
was employed for preparation of calibration standards
and quality control samples. Each calibration standard
contained lysate from approximately 50,000 MCF-10A
cellsandwaspreparedbyadding2μloftheSI-EM
working internal standard solution (16 pg of each SI-
EM) to various volumes of the EM working standard
solution. These calibration standards typically contain
0.2 to 200 pg of each EM in 0.5 ml of MCF-10A cell
lysate and were assayed in duplicate. The calibration
standards cover three orders of magnitude.
Sample preparation procedure
Samples were prepared and analyzed following a pre-
viously published method [20,21]. Briefly, each tumor
cell pellet contained approximately 1 million cells. They
were first suspended in 2 ml ice cold 12.5 mM
NH
4
HCO
3
solution. Cell lyses were prepared by tip
sonication on ice in five cycles of 10-second pulses and
10-second breaks followed by 30-minute water bath
sonication. To 0.5 ml of each cell lysate, 0.5 ml o f
freshly prepared 0.15 M sodium acetate buffer (pH 4.6)
containing 16 pg of each SI-EM and 2 mg of L-ascorbic
acid was added. Sa mples then underwent slow inverse
extraction at 8 rpm (RKVSD,ATR,Inc.,Laurel,MD,
USA) with 5 ml dichloromethane for 30 minutes. After
extraction, the organic solvent p ortion was transferred
into a clean glass tube and evaporated to dryne ss at 60°
C under nitrogen gas (Reacti-Vap III, Pierce, Rock-
ford, IL, USA).
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 2 of 11
To each dried sample, 32 μl of 0.1 M sodium acetate
buffer (pH at 9.0) and 32 μl of dansyl c hloride s olution
(1 mg/ml in acetone) were added. After vortexing, the
samplewasheatedat60°C(Reacti-ThermIII Heating
Module, Pierce, Rockford, IL, USA) for 10 minutes to
form the EM and SI-EM dansyl derivatives (EM-Dansyl
and SI-EM-Dansyl, respectively). Calibration standards
and quality control samples were hydrolyzed, extracted,
and derivatized following the same procedure used for
unknown cell samples. After derivatization, all samples
were analyzed by capillary liquid chromatography (LC)-
tandem mass spectrometry (MS
2
).
Liquid chromatography-tandem mass spectrometry
LC-MS
2
analysis was performed using an Agilent 1200
series nanoflow LC system (Agilent Technologies, Palo
Alto, CA, USA) coupled to a TSQ Quantum Ultra tri-
ple quadrupole m ass spect rometer ( Thermo Electron,
San Jose, CA, USA). The LC separation was carried out
on a 150 mm long × 300 μm internal diameter column
packed with 4 μm Synergi Hydro-RP particles (Phenom-
enex, Torrance, CA, USA) and maintained at 40°C. A total
of 8 μl of each sample was injected onto the column. The
mobile phase, operating at a flow rate of 4 μl/minute, con-
sists of methanol as solvent A and 0.1% (v/v) formic ac id
in water a s solvent B. A lin ear gradient from 72 to 85%
solvent B in 75 minutes was employed for separation of
EMs and SI-EMs. The mass spectrometry conditions were:
source, ESI; ion polarity,positive; spray voltage, 3200 V;
sheath and auxiliary gas, nitrogen; sheath gas pressure, 10
arbitrary units; ion transfer capillary temperature, 270 °C;
scan type, selected reaction monitoring; collision gas,
argon; collision gas pressure, 1.5 mTorr; scan width, 0.7 u;
scan time, 0.30 s; Q1 peak width, 0.70 u full-width half-
maximum (FWHM ); Q3 peak width, 0.70 u FWHM. The
optimized selected reaction monitoring conditions for the
protonated molecules [MH]
+
of EM-Dansyl and SI-EM-
Dansyl were similar to those previously described [9,10].
Quantification of estrogen metabolites
Quantification of EMs was carried out using Xcalibur
Quan Browser (Thermo Electron) as previously described
[20,21]. Brie fly, calibration curves for the each EM w ere
constructed by plotting EM-Dansyl/SI-EM-Dansyl peak
area ratios obtained from calibration standards versus
amounts of the EM injected on the column and fitting
these data using linear regression with 1/X weighting. The
amounts of EMs in cells were then interpolated using this
linear function. Based on their similarity of structures and
retention times,
13
C
6
-E
2
was used as the internal standard
for E
2
;
13
C
6
-E
1
for E
1
;d
3
-E
3
for E
3
, 16-ketoE
2
,and16a-
OHE
1
;d
3
-16-epiE
3
for 16-epiE
3
and 17-epiE
3
;d
5
-2-
MeOE
2
for 2-MeOE
2
, 4-MeOE
2
, 2-MeOE
1
, 4-MeOE
1
,and
3-MeOE
1
;d
5
-2-OHE
2
for 2-OHE
2
, 2-OHE
1
, and 4-OHE
1
.
Results and discussion
The levels of endogenous estrogens and EMs were mea-
sured in the NCI-60 cell lines, which comprise breast (n =
5), CNS (n = 6), colon (n = 7), leukemia (n = 6), melanoma
(n = 9), non-small cell lung (n = 9), ovarian (n = 9), pros-
tate (n = 2), and renal (n = 8) cancers. This study focused
on measuring only the unconjugated, active forms o f the
EMs. Glucoronidated and sulfated forms of the EMs were
not included in the analysis. All NCI-60 human tumor cell
lines showed significant levels of E
1
,E
2
, 16-ketoE
2
,16a-
OHE
1
,E
3
,2-MeOHE
1
,2-MeOHE
2
,and2-OHE
1
.The
chromatograms showing the eight quantified endogenous
EMs for an ovarian (SK-OV-3) and colon cance r cell line
(HCC-2998) are shown in Figure 1. The peaks were gener-
ally well resolved and had good signal-to-noise ratios for
all cell lines analyzed. While undetectable in all the others,
2-OHE
1
was found i n the non-small cell lung cancer cell
line NCI-H460.
Within the same type of cancer, different tumor cell
lines had substantially different levels o f EMs (Table 1).
For example, SF-539 and SNB-75 cells produced greater
amounts of estrogens than the other CNS lines tested.
HCC-2998 colon ca ncer cells, RMPI-8226 leukemia
cells, SK-MEL-28, UACC-257, UACC-62, MALME-3M
melanoma cells, EKVX, NCI-H23, NCI-H226 non-small
cell lung (NSCL) cancer cells, OVCAR-4, OVCAR-5,
SK-OV-3 ovarian cancer cells, and CAKI-1 renal ca ncer
cells all produced greater amou nts of estrogens than the
other cell line s within thei r category. Furthermore,
estrogen levels in these tumor cell lines were substan-
tially elevated and even exceeded the levels typically
found in well characterized estrogen-dependent and ER-
positive tumor cells such as MCF-7 and T-47D.
Within each tumor cell line, E
2
was by far the most
abundant unconjugated estrogen followed by E
1
and 2-
OHE
2
(Table 1). For the five b reast cancer cell lines, E
2
represented 75 to 85% of the tot al amount of unconju-
gated EMs measured. For six of the ovarian cancer cell
lines, E
2
represented 77 to 87% of the total estrogen con-
tent, while this percentage was only 62% for OVCAR-3
cells. T-47D and MCF-7 cells are both estrogen-dependent
and ER-positive human breast cancer cells and have E
2
levels at 87 and 81 pg/10
6
cells, which accounted for 85%
and 82% of their total unconjugated estrogens, respe c-
tively. MDA-MB-231 is an estroge n-independent, ER-
negative, HER2-positive human breast cancer cell line and
still has E
2
levels of about 37 pg/10
6
cells, which accounts
for about 75% of its total unconjugated estrogen levels.
The cell lines with the highest E
2
levels are shown in
Figure 2a. Although estrogens are commonly associated
with breast cancer, none of these cel l lines were among
those that contained the highes t levels of E
2
. Co nsistent
with evidence linking estrogen level s with cancers of the
reproductive system in general, three ovarian canc er cell
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 3 of 11
lines (OVCAR- 4, OVCAR-5, and SK-OV-3) were
amongst thos e with the highest E
2
levels. OVCAR-4 and
-5 are both ERa-negative, ERb-positive ovarian cell lines
whosgrowthisinsensitivetoE
2
treatment [22]. While
SK-OV-3 cells do express ERa, their growth is also
insensitive to treatment with E
2
[22]. The leukemia cell
line RMPI-8226 possessed the highest levels of E
2
(753
pg/10
6
cells). In fact, its E
2
levels were more than 3.5-
fold higher than the colon cell line HCC-2998, which
contained the next highest level of E2 (209 pg/10
6
cells).
This result correlates with a prev ious study showi ng
that RMPI-8226 cells possess the highest ER levels com-
pared to other leukemia and myeloid cell lines tested
[23]. Previous studies have shown that the HL60 leuke-
mia cell line possesses ERs and its proliferation is sensi-
tive to E
2
treatment. When the cells are maintained in
a medium containing physiological concentrations
(10
-9
M, 10
-8
M, 10
-7
M) of E
2
, cell growth is stimulated;
however, pharmacological concentrations (10
-6
M) of E
2
inhibit their growth [24]. Adding tamoxifen inhibited
the stimu lating effect of the est rogens by binding to and
blocking the ER. The effect of estrogen was therefore
associated with the presence of ERs in the human leuke-
mic cell line HL60 and may be important in the prolif-
eration of other leukemic cell lines.
Cell lines with the lowest E
2
levels are shown in
Figure 2b. Four of these were colon cell lines (HCT-116,
HCT-15, KM12, and SW-620). Their E
2
levels ranged
from 1.31 to 12.25 p g/10
6
cells. To put into perspective
the range of E
2
values found in all the cell lines, SW-
620 colon cells contained almost 575- fold less E
2
than
RMPI-8226 cells. The finding that colon cell lines gener-
ally contain low levels of E
2
is consistent with a previous
study that found ERs are present in colorectal tumors
and human colonic cancer cell lines at very low levels
[25].
Figure 1 Chromatograms showing the eight quantified endogenous estrogen metabolites for (a) the ovarian cancer cell line SK-OV-3
and (b) the colon cancer cell line HCC-2998. 16-ketoE
2
, 16-ketoestradiol; 16a-OHE
1
,16a-hydroxyestrone; 2-MeOE
1
, 2-methoxyestrone; 2-
MeOE
2
, 2-methoxyestradiol; 2-OHE
2
, 2-hydroxyestradiol; E
1
, estrone; E
2
, estradiol; E
3
, estriol.
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 4 of 11
Table 1 Levels of unconjugated endogenous estrogens (picograms) found in NCI-60 cell lines
Cancer Cell Line E
3
16aOHE
1
16-epiE
3
2MeOE
1
2MeOE
2
E
1
E
2
2OHE
1
2OHE
2
Total
Breast T-47D 0.80 0.03 0.33 2.70 0.54 3.65 87.37 NF 7.30 102.72
(0.07) (0.00) (0.04) (0.34) (0.10) (0.06) (2.69) NF (1.22) (3.18)
Breast Hs-578T 0.43 0.03 0.22 0.62 0.94 8.13 74.11 NF 5.86 90.35
(0.06) (0.00) (0.05) (0.07) (0.17) (0.98) (9.94) NF (0.83) (11.95)
Breast BT-549 0.58 0.02 0.06 0.65 0.45 8.45 58.97 NF 6.03 75.22
(0.10) (0.00) (0.03) (0.10) (0.02) (1.20) (11.51) NF (1.26) (14.15)
Breast MDA-MB-231 0.90 0.02 0.57 1.78 0.35 6.30 36.73 NF 2.55 49.21
(0.07) (0.00) (0.05) (0.24) (0.05) (0.37) (2.41) NF (0.33) (2.71)
Breast MCF-7 0.40 0.04 0.28 2.12 5.38 7.70 80.75 NF 1.76 98.42
(0.03) (0.00) (0.06) (0.23) (0.47) (0.71) (14.44) NF (0.06) (13.04)
CNS SF-295 0.24 0.04 0.12 0.93 0.81 6.79 41.51 NF 7.01 57.44
(0.03) (0.00) (0.01) (0.07) (0.12) (0.93) (2.57) NF (1.03) (3.25)
CNS SF-539 0.82 0.11 NF 1.73 1.22 26.24 175.50 NF 27.11 232.78
(0.10) (0.02) NF (0.13) (0.08) (2.01) (13.14) NF (2.78) (14.43)
CNS SF-268 0.35 0.04 0.31 0.43 0.26 2.69 24.68 NF 7.67 36.42
(0.01) (0.00) (0.03) (0.05) (0.01) (0.10) (1.33) NF (0.96) (2.21)
CNS U251 0.35 0.29 0.21 1.19 0.31 4.54 41.06 NF 12.61 60.55
(0.04) (0.03) (0.02) (0.11) (0.04) (0.21) (4.27) NF (1.22) (5.01)
CNS SNB-19 0.31 0.02 0.08 1.27 0.55 5.09 29.34 NF 3.63 40.29
(0.05) (0.00) (0.01) (0.14) (0.09) (0.64) (2.31) NF (0.25) (3.45)
CNS SNB-75 0.30 0.04 0.10 1.46 0.56 13.15 94.94 NF 2.23 112.79
(0.04) (0.00) (0.01) (0.23) (0.03) (0.57) (1.97) NF (0.24) (1.80)
Colon SW-620 0.75 0.08 0.28 1.70 1.39 0.44 1.31 NF 2.68 8.63
(0.11) (0.01) (0.05) (0.14) (0.23) (0.05) (0.09) NF (0.31) (0.61)
Colon HCT-116 0.12 0.02 0.27 1.40 0.80 0.45 2.17 NF 0.33 5.58
(0.02) (0.00) (0.04) (0.06) (0.08) (0.04) (0.13) NF (0.03) (0.11)
Colon COLO-205 0.33 0.07 0.19 0.57 0.76 1.12 50.44 NF 15.20 68.69
(0.02) (0.01) (0.02) (0.07) (0.07) (0.08) (4.57) NF (0.77) (5.22)
Colon KM12 0.55 0.04 0.31 1.09 0.44 0.14 1.77 NF 0.73 5.06
(0.06) (0.00) (0.04) (0.10) (0.09) (0.01) (0.12) NF (0.13) (0.49)
Colon HT29 0.51 0.03 0.51 1.12 0.28 1.90 30.71 NF 0.51 35.56
(0.01) (0.00) (0.06) (0.12) (0.03) (0.10) (1.12) NF (0.05) (0.90)
Colon HCT-15 28.10 0.40 1.08 0.87 0.29 0.88 12.25 NF 3.75 47.62
(1.54) (0.06) (0.04) (0.09) (0.03) (0.10) (1.22) NF (0.36) (3.05)
Colon HCC-2998 0.88 0.10 0.45 3.10 0.92 17.10 209.34 NF 4.29 236.18
(0.05) (0.01) (0.07) (0.05) (0.17) (0.08) (13.95) NF (0.14) (14.05)
Leukemia HL-60 3.89 0.03 0.29 1.58 3.15 18.78 36.61 NF 16.50 80.83
(0.02) (0.00) (0.01) (0.11) (0.10) (2.39) (2.70) NF (2.81) (2.39)
Leukemia CCRF-CEM 0.41 0.04 0.06 0.66 1.33 1.18 3.48 NF 1.16 8.31
(0.07) (0.00) (0.02) (0.04) (0.10) (0.12) (0.50) NF (0.10) (0.91)
Leukemia K562 0.42 0.02 0.21 0.57 2.23 0.56 13.88 NF 1.16 33.14
(0.07) (0.00) (0.04) (0.06) (0.07) (0.06) (1.75) NF (0.11) (1.24)
Leukemia MOLT-4 0.30 0.08 0.13 0.55 0.77 2.04 7.77 NF 0.75 12.37
(0.04) (0.01) (0.01) (0.04) (0.07) (0.37) (1.14) NF (0.09) (1.53)
Leukemia RMPI-8226 0.93 0.11 0.59 2.04 3.60 182.06 753.45 NF 2.93 945.71
(0.07) (0.02) (0.11) (0.30) (0.16) (5.19) (48.20) NF (0.22) (52.18)
Leukemia SR 4.67 0.06 0.80 3.24 1.45 6.67 26.39 NF 1.22 44.51
(0.53) (0.01) (0.15) (0.51) (0.22) (0.47) (1.38) NF (0.19) (2.00)
Melanoma MDA-MB-435 0.37 0.22 0.09 0.61 3.65 1.77 14.24 NF 2.82 23.77
(0.03) (0.04) (0.02) (0.11) (0.76) (0.18) (0.91) NF (0.32) (1.05)
Melanoma SK-MEL-28 0.82 0.05 0.27 3.83 5.41 21.50 146.59 NF 27.77 206.24
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 5 of 11
Table 1 Levels of unconjugated endogenous estrogens (picograms) found in NCI-60 cell lines (Continued)
(0.03) (0.01) (0.02) (0.33) (0.99) (2.12) (15.34) NF (2.17) (16.78)
Melanoma UACC-257 1.20 0.36 0.37 0.74 7.00 36.36 130.70 NF 2.50 179.23
(0.23) (0.07) (0.05) (0.10) (0.78) (2.41) (4.37) NF (0.22) (6.37)
Melanoma LOX IMVI 0.93 0.07 0.44 1.33 1.01 0.35 12.10 NF 8.26 24.49
(0.11) (0.01) (0.07) (0.06) (0.07) (0.02) (0.55) NF (0.64) (0.50)
Melanoma UACC-62 0.20 0.30 0.06 1.41 1.47 17.49 98.71 NF 1.62 121.27
(0.03) (0.04) (0.01) (0.06) (0.25) (0.21) (4.61) NF (0.30) (4.82)
Melanoma SK-MEL-2 1.56 1.60 0.38 2.21 0.62 6.66 50.93 NF 2.90 66.86
(0.15) (0.08) (0.07) (0.05) (0.08) (0.40) (5.84) NF (0.33) (6.40)
Melanoma SK-MEL-5 0.38 0.38 0.13 14.84 0.52 11.64 72.02 NF 3.83 103.75
(0.03) (0.05) (0.02) (0.47) (0.08) (0.31) (4.06) NF (0.50) (5.25)
Melanoma MALME-3M 0.36 0.06 0.32 3.88 0.45 1.26 94.27 NF 1.30 101.90
(0.03) (0.01) (0.05) (0.26) (0.06) (0.05) (0.88) NF (0.13) (1.13)
Melanoma M14 1.33 0.05 0.18 1.24 0.69 1.21 13.95 NF 1.90 20.55
(0.06) (0.00) (0.02) (0.11) (0.11) (0.09) (0.50) NF (0.28) (0.91)
NSCL A549 0.56 0.04 0.54 1.04 1.18 2.95 17.01 NF 2.19 25.51
(0.06) (0.00) (0.05) (0.05) (0.07) (0.27) (0.86) NF (0.24) (1.18)
NSCL EKVX 1.70 0.31 0.92 1.10 7.16 15.16 125.95 NF 4.53 156.83
(0.06) (0.05) (0.01) (0.12) (0.72) (1.13) (13.92) NF (0.47) (12.21)
NSCL HOP-62 1.48 0.08 0.78 2.37 4.70 6.19 25.45 NF 13.29 54.33
(0.24) (0.01) (0.14) (0.40) (0.44) (1.18) (1.85) NF (1.56) (5.04)
NSCL NCI-H23 1.08 0.29 0.39 6.10 0.92 8.03 90.47 NF 15.79 123.07
(0.05) (0.04) (0.08) (0.43) (0.05) (0.64) (2.81) NF (2.87) (3.46)
NSCL NCI-H460 0.18 0.05 0.15 1.36 1.15 4.03 59.76 27.89 2.21 96.78
(0.01) (0.01) (0.00) (0.23) (0.04) (0.06) (3.33) (0.32) (0.12) (3.69)
NSCL NCI-H226 0.29 0.07 0.13 0.50 0.61 25.40 181.13 NF 21.63 229.75
(0.02) (0.01) (0.01) (0.02) (0.07) (1.05) (7.48) NF (0.69) (8.87)
NSCL HOP-92 0.33 0.03 0.43 0.55 0.24 4.87 34.58 NF 2.20 43.24
(0.01) (0.00) (0.08) (0.03) (0.04) (0.28) (1.51) NF (0.24) (1.43)
NSCL NCI-H522 0.18 0.03 0.06 1.38 0.31 4.25 32.36 NF 1.55 40.12
(0.02) (0.00) (0.01) (0.08) (0.04) (0.36) (2.39) NF (0.31) (2.58)
NSCL NCI-H322M 0.19 0.04 0.13 0.80 0.45 0.72 7.01 NF 1.29 10.62
(0.02) (0.00) (0.01) (0.06) (0.04) (0.08) (0.27) NF (0.12) (0.13)
Ovarian OVCAR-3 0.50 0.07 0.34 0.97 3.70 5.37 26.60 NF 5.47 43.02
(0.05) (0.01) (0.04) (0.09) (0.54) (0.73) (1.63) NF (0.81) (3.21)
Ovarian OVCAR-5 0.16 0.03 0.13 1.26 0.33 12.85 107.47 NF 1.57 123.79
(0.00) (0.00) (0.02) (0.17) (0.05) (0.33) (5.55) NF (0.14) (5.89)
Ovarian IGR-OV1 0.15 0.04 0.11 0.73 2.54 4.00 29.72 NF 0.66 37.94
(0.01) (0.00) (0.00) (0.01) (0.36) (0.15) (0.70) NF (0.06) (1.10)
Ovarian NCI/ADR-RES 1.33 0.15 0.44 2.54 2.92 10.46 75.10 NF 4.61 97.56
(0.08) (0.02) (0.04) (0.28) (0.14) (0.31) (0.47) NF (0.28) (0.06)
Ovarian SK-OV-3 1.78 0.13 0.39 0.73 0.55 12.26 132.33 NF 4.18 152.36
(0.23) (0.02) (0.03) (0.09) (0.10) (0.25) (9.43) NF (0.63) (9.94)
Ovarian OVCAR-8 0.44 0.13 0.15 2.10 0.35 5.12 38.83 NF 3.61 50.73
(0.02) (0.03) (0.03) (0.17) (0.06) (0.22) (1.01) NF (0.58) (1.56)
Ovarian OVCAR-4 0.43 0.06 0.31 3.90 0.43 19.79 139.32 NF 4.69 168.93
(0.02) (0.01) (0.02) (0.06) (0.07) (1.06) (2.84) NF (0.46) (2.82)
Prostate PC-3 0.15 0.03 0.20 1.05 2.33 7.63 41.94 NF 0.63 53.96
(0.02) (0.00) (0.03) (0.05) (0.14) (0.56) (2.30) NF (0.07) (2.90)
Prostate DU145 0.55 0.04 0.42 2.22 0.71 4.94 27.78 NF 2.55 39.22
(0.05) (0.00) (0.05) (0.16) (0.10) (0.29) (1.02) NF (0.23) (1.06)
Renal UO-31 0.30 0.13 0.33 1.36 1.66 5.05 22.94 NF 2.45 34.22
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 6 of 11
As with E
2
, the leukemic cell line RPMI-8226 con-
tained the highest levels of E
1
of the NCI-60 cell lines
(Figure 3a). The amount measured in this cell line was
more than five-fold higher than that f ound in the cell
line (melanoma UACC-257) cont aining the ne xt highest
levels of E
1
. Again, none of the five brea st ca ncer cell
lines tested was amongst the top ten E
1
-containing cells.
The ovarian cancer cell line OVCAR-4 (19.79 pg/10
6
cells) was sixth on the list of cell lines containing the
most E
1
. Two non-sma ll cell lung carci noma cell l ines
(EKVX and NCI-H226) were amongst the top ten in
both E
1
and E
2
levels. This result is interesting consider-
ing that females that never smoked are far more likely
to develop lung carcinoma than never-smoked males,
suggesting a gende r differenc e exists in the clinical and
pathophysiology of lung cancer [21]. Recent studies
showing aromatase-dependent synthesis of estrogens in
situ in male and female lun g cancers sugges t that estro-
gens may contribute to the manifestation and progres-
sion of lung carcinoma [26-28].
Cell lines with the lowest E
1
levels are shown in
Figure 3b. COLO-205 was included along with the four
colon cancer cell lines that were amongst the ten con-
taining the lowest E
1
levels (HCT-116, HCT-15, KM12,
and SW-620). Their E
1
levels ranged from 0.14 to 1.12
pg/10
6
cells. The levels of E
1
found in KM12 colo n can-
cer cells was approximately 1,300-fold less than that
found in RPMI-8226 leukemia cells. T wo leukemia cell
lines, CC RF-CE M and K562, whic h were amongst those
possessing the lowest E
2
levels, also contained low E
1
levels.
To identify general trends within the various cell lines
tested, the means and standard deviations (SDs) of the
total EM levels found in the cell types analyzed. As
shown in Table 2, the leukemia cel l lines had the hi ghest
overall mean total EM values ( 187.5 pg/10
6
cells). This
value was almost twice as h igh as the cell types with the
next highest total EM levels. Ovarian (96.33 pg/10
6
cells)
andbreast(83.18pg/10
6
cells) cancer cells, which are
commonly associated with estrogens, c ontained the sec-
ond and sixth highest levels of total EMs. In fact, five of
the cancer cell lines (ovarian, melanoma, CNS, NSCL,
and breast) had t otal EM levels between 80 and 100 pg/
10
6
cells. A noticeable feature of Table 2 are the very
highSDs,butinparticularthatfortheleukemiacell
lines. T o further explore what contributed to this high
SD, the highest and lowest total EM levels measure d for
the individual cell lines in the different cell types w ere
eliminated and the mean and SD values were recalcu-
lated. The mea n E M levels for the leukemia cell lines
after eliminating the cell lines with the highest and lowest
concentrations was 42.71 pg/10
6
cells with a SD of 28.68;
dropping their overall rank from first to sixth. The cell
types having the three highest EM levels after eliminating
the cell lines with the highest and lowest concentrations
were (in order) ovarian (93.29 pg/10
6
cells, SD = 46.84),
melanoma (88.75 pg/10
6
cells, SD = 55.53), and breast
(88.00 pg/10
6
cells, SD = 11.78).
The finding that the melanoma cell lines contained
relatively high levels of endogenous estro gen and EMs is
interesting. Two melanoma cell lines in particular, SK-
MEL-28 and UACC-257, were among those containing
the highest levels of E
1
(21.50 a nd 36.36 pg/10
6
cells,
respectively) and E
2
(146.59 and 130.70 pg/10
6
cells,
respectively). Only four other cell lines, SF-539 (CNS),
NCI-H226 (NSCL), RMPI-8226 (leukemia) and HCC-
2998 (colon), contained h igher levels of total estrogens.
High-affinity E
2
receptors have been reported for primary
Table 1 Levels of unconjugated endogenous estrogens (picograms) found in NCI-60 cell lines (Continued)
(0.03 (0.02 (0.02 (0.26 (0.26 (0.60 (1.18 NF (0.15 (0.92
Renal 786-0 0.15 0.02 0.11 0.58 0.16 2.11 23.40 NF 1.54 28.07
(0.02) (0.00) (0.01) (0.05) (0.02) (0.16) (0.14) NF (0.19) (0.17)
Renal SN12C 0.14 0.05 0.27 6.47 1.92 4.02 25.50 NF 0.89 39.25
(0.01) (0.01) (0.02) (0.29) (0.17) (0.07) (0.97) NF (0.05) (0.96)
Renal CAKI-1 0.54 0.16 0.13 4.76 0.49 7.57 81.60 NF 6.52 101.77
(0.05) (0.03) (0.03) (0.42) (0.07) (0.61) (1.42) NF (0.79) (2.29)
Renal RXF-393 0.27 0.03 0.09 0.47 0.44 0.34 14.48 NF 1.41 17.54
(0.04) (0.00) (0.02) (0.09) (0.04) (0.04) (0.39) NF (0.13) (0.38)
Renal TK-10 0.65 0.25 0.42 2.54 0.58 7.87 37.29 NF 6.25 55.86
(0.06) (0.04) (0.02) (0.24) (0.06) (0.22) (3.96) NF (0.60) (4.27)
Renal A498 0.53 0.02 0.16 0.29 0.50 3.05 37.53 NF 2.58 44.65
(0.04) (0.00) (0.00) (0.03) (0.07) (0.29) (1.09) NF (0.37) (1.45)
Renal ACHN 0.43 0.08 0.19 1.25 0.39 6.17 39.27 NF 3.40 51.17
(0.06) (0.01) (0.02) (0.16) (0.06) (0.74) (1.96) NF (0.28) (2.51)
Data are expressed as mean (standard deviation) of three replicated analyses of 10
6
cells. E
3
, estriol; 16aOHE
1
,16a-hydroxyestrone; 16-epiE
3
, 16-epiestriol;
2MeOE
1
, 2-methoxyestrone; 2MeOE
2
, 2-methoxyestradiol; E
1
, estrone; E
2
,17b-estradiol; 2OHE
1
, 2-hydroxyestrone; 2OHE
2
, 2-hydroxyestradiol; CNS, central nervous
system; NF, not found; NSCLC, non-small cell lung carcinoma.
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 7 of 11
human melanomas [29] and patients expressing these
receptors seem to have a better prognosis, suggesting E
2
may inhibit the growth of t hese m elanoma t umors [30].
While previous studies have identified the cl assical ER in
only a small percentage of human melanomas via immu-
nohistochemistry [30], the low affinity type II ER has
been characterized in a variety of human melanomas
[31]. This receptor has the same affinity as the classical
receptor and also binds tamoxifen. Treating SK-Mel 23
melanoma cells with E
2
has been shown to inhibit their
growth, while pre-treating the cells with tamoxifen (an
anti-estrogen) blocks the effects of E
2
[32]. The preva-
lence of E
2
within the melanoma cell lines may prevent
uncontrolled cell proliferation by acting back upon the
cells and binding to the type II ER.
In general, unconjugated E
3
,16aOHE
1
, and 16-epiE
3
were less abundant except for in HCT-15 colon tumor
cells, which had a greater amount of E
3
than E
2
.The
catechol estrogen 2-OHE
2
was the only catechol estro-
gen detected in the tumor cell lines, except for the
NSCL cancer cell line NCI-H460, which also contains a
relatively high level of 2-OHE
1
(Table 1). N o unconju-
gated 4-hydroxy catechol estrogens were detected in any
of the NCI-60 tumor cells. This result is likely due to
the fa ct that 4-hydroxy catechol estrogens are quickly
transformed into other reactive species such as their
quinones and semi-qui nones, which could damage DNA
and l ead to tumor initiation [2,33,34]. In contrast, 2-
hydroxy-catechol estrogens largelyformstableconju-
gates such as 2-MeOHE
1
and 2 -MeOHE
2
. S ignificant
Figure 2 Cell lines containing the highest and lowest 17b-estradiol (E
2
) levels within the NCI-60 panel. (a) Cell lines with the highest E
2
levels; (b) cell lines with the lowest E
2
levels.
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 8 of 11
Figure 3 Cell lines containing the highest and lowest estrone (E
1
) levels within the NCI-60 panel. (a) Cell lines with the highest E
1
levels;
(b) cell lines with the lowest E
1
levels.
Table 2 Means and standard deviations of total estrogen levels measured in cell types within the NCI-60 cell line
panel
Mean SD Mean (-high/low)* SD (-high/low)*
Breast 83.18 21.70 88.00 11.78
CNS 90.05 75.07 67.77 31.31
Colon 58.19 82.12 33.21 26.64
Leukemia 187.48 372.4 42.71 28.67
Melanoma 94.23 67.69 88.75 55.49
NSCL 86.69 71.98 77.13 49.25
Ovarian 96.33 54.00 93.49 46.84
Prostate 46.59 10.42 NA NA
Renal 46.57 25.49 42.20 10.44
Mean and standard deviation (SD) are presented for all values within specific cell types and for values after eliminating cell lines with the highest and lowest
total estrogen levels. CNS, central nervous system; NSCLC, non-small cell lung carcinoma. *Mean and standard deviation calculated after removing cell lines
containing highest and lowest total measured estrogen levels. NA, only two prostate cell lines were included in NCI-60 cell lines analyzed.
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 9 of 11
levels of b oth of these EMs were found in all NCI-60
cell lines tested in this study.
This study measured the unconjugated levels o f endo-
genous estrogens and EMs in the NCI-60 cell panel. From
our previous experience, if we had measured the conju-
gated levels by adding a sulfatase/glucoronidase enzyme to
deconjugat e sulfat ed and glucoronida ted molecules prior
to LC-MS
2
analysis, we would expect to see a large
increase in the levels of every metabolite that was routinely
detected. We would also expect that 16-epiE
3
,17-epiE
3
,4-
MeOE
1
, 3-MeOE
1
, 4-MeOE
2
,2-OHE
1
, and 4-OHE
1
would
also be detectable. Our studies analyzing serum have
shown that endogenous estrogens and EMs exist primarily
(that is, 90%) in the conjugated forms in the circulation
[20]. While this disparity between conjugated and uncon-
jugated forms of these steroid hormones may not be as
large i n cells, we predict that a large amou nt of endogen-
ous estrogens and EMs exist within cells in their conju-
gated forms. It is interesting to note that when we analyze
serum, only E
1
,E
2
,E
3
,2-MeOHE
1
and 2-MeOHE
2
are
detected in their uncon jugated forms [20]. In th e NCI-60
cell lines we were able to also routinely detect 16-aOHE
1
,
16-epiE
3
,and2-OHE
2
. Unfortunately, it is difficult to
directly compare the estrog en and EM levels as the cell
line concentrations are recorded in pg/10
6
cells, while
those in serum are measured as pg/ml. The fact that more
compounds are detected in their unconjugated forms in
the cell lines, however, sugge sts that, in general, the con-
centrations of estrogens and EMs are higher in cells than
in the circulation.
To determine if ER status correlates with the levels of
estrogensandEMsidentifiedinthevariouscelllines,
we compare our data to that published by Holbeck et
al. [35], who measured the mRNA levels of 48 nuclear
receptors in 51 of the NCI-60 cell lines. The mRNA
levels of ERa for nine of the cell lines that were found
to contain the highest E
2
levels were measured in this
study. Of these, detecta ble ER a levels were found f or
SKOV-3, OVCAR-4, UACC-257, SK-MEL-28, and SF-
539 cell lines. No ERa mRNA was detected f or H CC-
2998, NCI-H226, EKVX, and OVCAR-5 cell lines. The
cell lines with the highest levels of ERa mRNA were
SK-OV-3, and two breast cancer cell lines, MCF-7 and
T-47D.Ofthese,onlySK-OV-3wasamongthecell
lines containing the highest amounts of E
2
.Wealso
compared the ERa and E
2
levels wit hin the nine mela-
noma cell lines analyzed in both studies. In this case,
the six melanoma cell lines with detectable levels of
ERa mRNA (SK-MEL-28, UACC-257, UACC-62, SK-
MEL-2, SK-MEL-5, and MALME-3M) contained the
highest amounts of E
2
within that group. The melanoma
cell lines containing the lowest E
2
concentrations (M14,
LOX IMVI, and MDA-MB-435) did not show detectable
levels of ERa. Overall, there is no obvious correlation
between ERa and E
2
levels; however, only about 25% of
the cell lines had detectable levels of ERa whereas E
2
could be measured in every one.
Conclusions
This study utilized an LC-MS
2
approach with the cap-
ability of measuring up t o 15 different EMs to measure
the l evels o f e ndogenous e strogens within the NCI-60
cell lines. Eight of the measured endogenous estrogens
were consistently observed in all of the NCI-60 cell
lines, providing an unprecedented view of these metabo-
lites within these cancer cell lines. What is particularly
striking is that the levels of EMs in well-known estro-
gen-dependent cancers such as o var ian and breast were
not substantially grea ter than those found in other types
of cancer cell lines. In fact, none of the breast cancer
cell lines were amongst the top ten that contained the
highest levels of E
1
or E
2
. Cell lines not generally asso-
ciated with estrogens, such as leukemia, colon, CNS,
and NSCL, were found to h ave a ppreciable levels of
these m etabolites. The broad presence of EMs within
the NCI-60 cell lines suggests that many cancers outside
of the reproductive system may respond to treatments
with anti-estrogens such as tamoxifen, toremifene, and
fulvestrant. Considering technologies for measuring
estrogen levels in biologicalsamplesisconsiderably
improved, it is now worth the effort to test various
tumors for the levels of these metabolites.
Abbreviations
16-epiE
3
: 16-epiestriol; 16-ketoE
2
: 16-ketoestradiol; 16α-OHE
1
:16α-
hydroxyestrone; 17-epiE
3
: 17-epiestriol; 2-MeOE
1
: 2-methoxyestrone; 2-MeOE
2
:
2-methoxyestradiol; 2-OHE
1
: 2-hydroxyestrone; 2-OHE
2
: 2-hydroxyestradiol; 3-
MeOE
1
: 2-hydroxyestrone-3-methyl ether; 4-MeOE
1
: 4-methoxyestrone; 4-
MeOE
2
: 4-methoxyestradiol; 4-OHE
1
: 4-hydroxyestrone; CNS: central nervous
system; E
1
: estrone; E
2
: estradiol; E
3
: estriol; EM: estrogen metabolite; ER:
estrogen receptor; LC: liquid chromatography; MS
2
: tandem mass
spectrometry; NSCL: non-small cell lung; SD: standard deviation; SI-EM: stable
isotope-labeled estrogen. Bre: breast; Col: colon; Leu: leukemia; Mel:
melanoma; Ovc: ovarian; Pros: prostate; Ren: renal.
Acknowledgements
This project has been funded in whole or in part with federal funds from
the National Cancer Institute, National Institutes of Health, under contract
N01-CO-12400. The content of this publication does not nece ssarily reflect
the views or policies of the Department of Health and Human Services, nor
does mention of trade names, commercial products, or organizations imply
endorsement by the United States Government.
Authors contributions
TV participated in the conception, design, and implementation of the
experiments, the analysis and interpretation of the data and drafting of the
manuscript. XX participated in the conception, design, and implementation
of the experiments, conducted the sample preparation and data acquisition
and participated in the analysis and interpretation of the data and drafting
of the manuscript. All authors have read and approved the final version of
the manuscript for publication.
Competing interests
The authors declare that they have no competing interest s.
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 10 of 11
Received: 11 January 2012 Revised: 4 April 2012
Accepted: 30 April 2012 Published: 30 April 2012
References
1. Eliassen AH, Hankinson SE: Endogenous hormone levels and risk of
breast, endometrial and ovarian cancers: prospective studies. Adv Exp
Med Biol 2008, 630:148-165.
2. Cavalieri EL, Rogan EG: Depurinating estrogen-DNA adducts in the
etiology and prevention of breast and other human cancers. Future
Oncol 2010, 6 :75-91.
3. Márquez-Garbán DC, Chen HW, Goodglick L, Fishbein MC, Pietras RJ:
Targeting aromatase and estrogen signaling in human non-small cell
lung cancer. Ann N Y Acad Sci 2009, 1155:194-205.
4. Siegfried JM, Hershberger PA, Stabile LP: Estrogen receptor signaling in
lung cancer. Semin Oncol 2009, 36:524-531.
5. Di Leo A, Messa C, Cavallini A, Linsalata M: Estrogens and colorectal
cancer. Curr Drug Targets Immune Endocr Metabol Disord 2001, 1:1-12.
6. Ellem SJ, Risbridger GP: Aromatase and regulating the estrogen:androgen
ratio in the prostate gland. J Steroid Biochem Mol Biol 2010, 118:246-251.
7. Hogan AM, Collins D, Baird AW, Winter DC: Estrogen and its role in
gastrointestinal health and disease. Int J Colorectal Dis 2009, 24:1367-1375.
8. Hojo Y, Murakami G, Mukai H, Higo S, Hatanaka Y, Ogiue-Ikeda M, Ishii H,
Kimoto T, Kawato S: Estrogen synthesis in the brainrole in synaptic
plasticity and memory. Mol Cell Endocrinol 2008, 290:31-43.
9. Giordano C, Montopoli M, Perli E, Orlandi M, Fantin M, Ross-Cisneros FN,
Caparrotta L, Martinuzzi A, Ragazzi E, Ghelli A, Sadun AA, dAmati G,
Carelli V: Oestrogens ameliorate mitochondrial dysfunction in Lebers
hereditary optic neuropathy. Brain 2011, 134:220-234.
10. Felty Q, Yoo C, Kennedy A: Gene expression profile of endothelial cells
exposed to estrogenic environmental compounds: implications to
pulmonary vascular lesions. Life Sci 2010, 86:919-927.
11. Merklinger-Gruchala A, Ellison PT, Lipson SF, Thune I, Jasienska G: Low
estradiol levels in women of reproductive age having low sleep
variation. Eur J Cancer Prev 2008, 17:467-472.
12. Liehr JG: Is estradiol a genotoxic mutagenic carcinogen?. Endocr Rev
2000, 21:40-54.
13. Santen RJ, Brodie H, Simpson ER, Siiteri PK, Brodie A: History of aromatase:
saga of an important biological mediator and therapeutic target. Endocr
Rev 2009, 30
:343-375.
14. Krahn-Bertil E, Dos Santos M, Damour O, Andre V, Bolzinger MA: Expression
of estrogen-related receptor beta (ERRβ) in human skin. Eur J Dermatol
2010, 20:719-723.
15. Plewig G, Cunliffe WJ, Binder N, Höschen K: Efficacy of an oral
contraceptive containing EE 0.03 mg and CMA 2 mg (Belara) in
moderate acne resolution: a randomized, double-blind, placebo-
controlled Phase III trial. Contraception 2009, 80:25-33.
16. Shu YY, Maibach HI: Estrogen and skin: therapeutic options. Am J Clin
Dermatol 2011, 12:297-311.
17. Koomen ER, Joosse A, Herings RM, Casparie MK, Guchelaar HJ, Nijsten T:
Estrogens, oral contraceptives and hormonal replacement therapy
increase the incidence of cutaneous melanoma: a population-based
case-control study. Ann Oncol 2009, 20:358-364.
18. Adami HO, Persson I, Hoover R, Schairer C, Bergkvist L: Risk of cancer in
women receiving hormone replacement therapy. Int J Cancer 1989,
44:833-839.
19. Kennelly R, Kavanagh DO, Hogan AM, Winter DC: Oestrogen and the
colon: potential mechanisms for cancer prevention. Lancet Oncol 2008,
9:385-391.
20. Xu X, Roman JM, Issaq HJ, Keefer LK, Veenstra TD, Ziegler RG: Quantitative
measurement of endogenous estrogens and estrogen metabolites in
human serum by liquid chromatography-tandem mass spectrometry.
Anal Chem 2007, 79:7813-7821.
21. Blonder J, Johann DJ, Veenstra TD, Xiao Z, Emmert-Buck MR, Ziegler RG,
Rodriguez-Canales J, Hanson JA, Xu X: Quantitation of steroid hormones
in thin fresh frozen tissue sections. Anal Chem 2008, 80:8845-8852.
22. ODonnell AJM, Macleod KG, Burns DJ, Smyth JF, Langdon SP: Estrogen
receptor-α mediates gene expression changes and growth response in
ovarian cancer cells exposed to estrogen. Endocr Relat Cancer 2005,
12:851-866.
23. Danel L, Vincent C, Rousset F, Klein B, Bataille R, Flacher M, Durie BGM,
Revillard JP: Estrogen and progesterone receptors in some human
myeloma cell lines and murine hybridomas. J Steroid Biochem 1988,
30:363-367.
24. Danel L, Cordier G, Revillard JP, Saez S: Presence of estrogen binding sites
and growth-stimulating effect of estradiol in the human myelogenous
cell line HL60. Cancer Res 1982, 42:4701-4705.
25. Hendrickse CW, Jones CE, Donovan IA, Neoptolemos JP, Baker PR:
Oestrogen and progesterone receptors in colorectal cancer and human
colonic cancer cell lines. Br J Surg 1993, 80:636-640.
26. Verma MK, Miki Y, Sasano H: Aromatase in human lung cancer. Steroids
2011, 76:759-764.
27. Suzuki T, Abe K, Suzuki S, Niikawa H, Iida S, Hata S, Akahira J, Mori K,
Evans DB, Kondo T, Yamada-Okabe H, Sasano H:
Intratumoral localization
of aromatase and interaction between stromal and parenchymal cells in
the non-small cell lung carcinoma microenvironment. Cancer Res 2010,
70:6659-6669.
28. Miki Y, Abe K, Suzuki S, Suzuki T, Sasano H: Suppression of estrogen
actions in human lung cancer. Mol Cell Endocrinol 2011, 340:168-174.
29. Stoica A, Hoffman M, Marta L, Voiculetz N: Estradiol and progesterone
receptors in human cutaneous melanoma. Neoplasma 1991, 38:137-146.
30. Miller JG, Gee J, Price A, Garbe C, Wagner M, MacNeil S: Investigation of
oestrogen receptors, sex steroids and soluble adhesion molecules in
progression of malignant melanoma. Melanoma Res 1997, 7:197-208.
31. Piantelli M, Maggiano N, Ricci R, Larocca LM, Capelli A, Scambia G, Isola G,
Natali PG, Ranelletti OF: Tamoxifen and quercetin interact with type II
estrogen binding sites and inhibit the growth of human melanoma
cells. J Inv Dermatol 1995, 105:248-253.
32. Sarti MSMV, Visconti MA, Castrucci AML: Biological activity and binding of
estradiol to SK-MEL 23 human melanoma cells. Braz J Med Biol Res 2004,
37:901-905.
33. Yager JD, Davidson NE: Estrogen carcinogenesis in breast cancer. N Engl J
Med 2006, 354:270-282.
34. Bulun SE, Chen D, Lu M, Zhao H, Cheng Y, Demura M, Yilmaz B, Martin R,
Utsunomiya H, Thung S, Su E, Marsh E, Hakim A, Yin P, Ishikawa H, Amin S,
Imir G, Gurates B, Attar E, Reierstad S, Innes J, Lin Z: Aromatase excess in
cancers of breast, endometrium and ovary. J Steroid Biochem Mol Biol
2007, 106:81-96.
35. Holbeck S, Chang J, Best AM, Bookout AL, Mangelsdorf DJ, Martinez ED:
Expression profiling of nuclear receptors in the NCI60 cancer cell panel
reveals receptor-drug and receptor-gene interactions. Mol Endocrinol
2010, 24:1287-1296.
doi: 10.1186/gm330
Cite this article as: Xu and Veenstra: Concentration of endogenous
estrogens and estrogen metabolites in the NCI-60 human tumor cell
lines. Genome Medicine 2012 4:31.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Xu and Veenstra Genome Medicine 2012, 4:31
http://genomemedicine.com/content/4/4/31
Page 11 of 11
... Dimethyl sulfoxide (0.1%) was used as a vehicle control. The effect of adding estradiol (250 pg/ml) on cell viability was also determined [28]. ...
... Neurosphere dimensions were measured using Leica Stereomicroscope in bright field setting. The effect of addition of estradiol (250 pg/ml) on the neurosphere growth was also determined [28]. ...
Article
Full-text available
PurposeThe DNA alkylating agent temozolomide (TMZ), is the first-line therapeutic for the treatment of glioblastoma (GBM). However, its use is confounded by the occurrence of drug resistance and debilitating adverse effects. Previously, we observed that letrozole (LTZ), an aromatase inhibitor, has potent activity against GBM in pre-clinical models. Here, we evaluated the effect of LTZ on TMZ activity against patient-derived GBM cells.Methods Employing patient-derived G76 (TMZ-sensitive), BT142 (TMZ-intermediately sensitive) and G43 and G75 (TMZ-resistant) GBM lines we assessed the influence of LTZ and TMZ on cell viability and neurosphere growth. Combination Index (CI) analysis was performed to gain quantitative insights of this interaction. We then assessed DNA damaging effects by conducting flow-cytometric analysis of ˠH2A.X formation and induction of apoptotic signaling pathways (caspase3/7 activity). The effects of adding estradiol on LTZ-induced cytotoxicity and DNA damage were also evaluated.ResultsCo-treatment with LTZ at a non-cytotoxic concentration (40 nM) reduced TMZ IC50 by 8, 37, 240 and 640 folds in G76, BT-142, G43 and G75 cells, respectively. The interaction was deemed to be synergistic based on CI analysis. LTZ co-treatment also significantly increased DNA damaging effects of TMZ. Addition of estradiol abrogated these LTZ effects.ConclusionsLTZ increases DNA damage and synergistically enhances TMZ activity in TMZ sensitive and TMZ-resistant GBM lines. These effects are abrogated by the addition of exogenous estradiol underscoring that the observed effects of LTZ may be mediated by estrogen deprivation. Our study provides a strong rationale for investigating the clinical potential of combining LTZ and TMZ for GBM therapy.
... 44,45 Moreover, the cellular association decreased significantly after estrone modification in ERα-negative cell lines A2780 and OVCAR-4, which was due to the attachment of the hydrophobic estrone ligand ( Figure 4C and Table S6(3)). 46,47 CLEM is a technique that combines the unique capabilities of light and electron microscopy by studying the same sample with both modalities sequentially. The confocal signal provides the region of interest and further overlaps with the electron microscopic images providing ultrastructural details of the sample. ...
Article
The estrone ligand is used for modifying nanoparticle surfaces to improve their targeting effect on cancer cell lines. However, to date, there is no common agreement on the ideal linker length to be used for the optimum targeting performance. In this study, we aimed to investigate the impact of poly(poly ethylene glycol methyl ether methacrylate) (PPEGMEMA) linker length on the cellular uptake behavior of polymer-coated upconverting nanoparticles (UCNPs). Different triblock terpolymers, poly(poly (ethylene glycol) methyl ether methacrylate)-block-polymethacrylic acid-block-polyethylene glycol methacrylate phosphate (PPEGMEMAx-b-PMAAy-b-PEGMP3: x = 7, 15, 33, and 80; y = 16, 20, 18, and 18), were synthesized with different polymer linker chain lengths between the surface and the targeting ligand by reversible addition-fragmentation chain transfer polymerization. The estrone ligand was attached to the polymer via specific terminal conjugation. The cellular association of polymer-coated UCNPs with linker chain lengths was evaluated in MCF-7 cells by flow cytometry. Our results showed that the bioactivity of ligand modification is dependent on the length of the polymer linker. The shortest polymer PPEGMEMA7-b-PMAA16-b-PEGMP3 with estrone at the end of the polymer chain was found to have the best cellular association behavior in the estrogen receptor (ER)α-positive expression cell line MCF-7. Additionally, the anticancer drug doxorubicin•HCl was encapsulated in the nanocarrier to evaluate the 2D and 3D cytotoxicity. The results showed that estrone modification could efficiently improve the cellular uptake in ERα-positive expression cell lines and in 3D spheroid models.
... Cytokines, including "adipokines", as well as leptin and adiponectin, have been studied as possible modulators of renal cell carcinoma development [17,18,19,20,21]. Estrogen production, which is increased within adipose tissue, may also be involved in this phenomenon [22,23]. ...
Article
BACKGROUND: information about the influence of abdominal adipose tissue on the prognosis and aggressiveness of renal cell carcinoma is scarce.OBJECTIVE: to identify and quantify the relationship between abdominal adipose tissue and histopathologic characteristics of renal cell carcinoma.MATERIALS AND METHODS: data on patients with renal cell carcinoma treated with partial or radical nephrectomy were retrospectively collected. Visceral adipose tissue and subcutaneous adipose tissue were measured on preoperative computerized tomography scans. Histopathologic (pathologic 2002 TNM classification, tumor size, nuclear grade, necrosis, and metastasis) and clinical (sex, age, body mass index) variables were assessed. Visceral adipose tissue and subcutaneous adipose tissue were adjusted by patient height (VAT-I and SAT-I). The association between adipose tissue measurements, patient characteristics, and histopathologic variables was assessed through a correlation, comparison, and regression analysis. A survival analysis was also carried out.RESULTS: one-hundred and five patients were included. No significant relationship between visceral adipose tissue and histopathologic characteristics was found. SAT-I was a significant variable inversely related to sex, but more importantly to nuclear grade, pN+, and necrosis. There was no influence of visceral adipose tissue or subcutaneous adipose tissue on survival.CONCLUSIONS: subcutaneous adipose tissue was related to the histopathologic characteristics of renal cell carcinoma. A larger amount of subcutaneous adipose tissue predicted a low nuclear grade and a lower frequency of pN+ and necrosis. Further study to clarify the interaction between adipose tissue and renal cell carcinoma biology is required.KEY WORDS: renal cell carcinoma, subcutaneous-abdominal adipose tissue, prognosis
... Here no additional bene t was obtained by testing in a speci c breast cancer cell line panel (32). This may be explained by the presence of oestrogen and derivates in all 60 cell lines with surprising abundancy in melanoma and leukaemia cell lines (33). ...
Preprint
Full-text available
Background Even with positive oestrogen receptor (ER+) status some advanced breast cancer (ABC) patients fail to benefit from endocrine therapy (ET). A method that previously predicted other drugs in various cancers was evaluated. Here multigene markers based on aromatase inhibitor (AI) effect in vitro were used for prediction of AI benefit in ER+ ABC patients. Simultaneously effects of long-term ET on predictive efficacy was evaluated. Methods The Drug Response Predictors (DRPs) are based on correlations between baseline gene expression and growth inhibition patterns of exemestane, anastrozole and letrozole, respectively, in the National Cancer Institute 60 cell lines. The genes were controlled for expression in 3,500 tumours. In a Danish Breast Cancer Cooperative Group cohort of 695 ABC patients with complete gene expression and time-to-progression (TTP) data, 414 received an AI as monotherapy. Hereof, 57 received anastrozole, 166 received exemestane, and 327 received letrozole. mRNA was isolated from archival formalin-fixed paraffin embedded tumour tissue and run on microarray and 60% of the tumours were from time of primary diagnosis. Medical records of the patients were assessed for TTP for all treatments given for ABC. Results The DRPs were tested in subsets 1) with no adjuvant ET and 2) with adjuvant ET. In 1) the anastrozole DRP predicted benefit of anastrozole (hazard ratio (HR) was 0.21 upper 95%-confidence interval limit (CI) 0.76, p=0.023) but not in 2). Dichotomised by a DRP of 50, the anastrozole DRP did predict benefit (HR=0.16, upper 95%-CI 0.75, p=0.026). Only in 1) the exemestane DRP predicted benefit of exemestane (HR=0.57, upper 95%-CI 1.00, p=0.0497). The letrozole DRP had no predictive value. Additionally, we tested each DRPs ability to predict other AIs. Only the anastrozole DRP predicted benefit of overall AI treatment, in 1) with an HR of 0.76 (upper 95%-CI 0.99, p=0.044) and in 2) with an HR of 0.71 (upper 95%-CI 0.92, p=0.015). The anastrozole DRP did though not predict benefit of letrozole. All tests are one-sided, alpha=5%. Conclusions Among the DRPs for AIs, the anastrozole DRP was strongest with clinically relevant prediction of TTP in AI treated ER+ ABC patients. Trial registration: ClinicalTrials.gov NCT01861496.
... Recent studies indicated that environmental estrogenic factors are involved in the progression of AML (Van Maele-Fabry et al., 2019). The estrogen receptor can regulate the proliferation of Hut-78 T cell and Jurkat cell (Xu and Veenstra, 2012). Phthalates, which have been identified as endocrine-disrupting chemicals (EDCs), can interfere the endocrine system to promote the development of various human diseases (Upson et al., 2013). ...
Article
Acute Myeloid Leukemia (AML) is a cancer of hematopoietic stem cells with a rapid progression. Recent studies indicated that endocrine disruptor chemicals (EDCs) are potential risk factors for AML progression. Our present data showed that an industrial endocrine disrupting chemical, Benzyl butyl phthalate (BBP), can promote the proliferation of AML cells and decrease their sensitivity to daunorubicin (DNR) and cytarabine (Ara-C) treatments. Further, BBP can increase the glucose consumption, lactate generation, and ATP levels of AML cells. Among the measured glycolysis-related genes, BBP can increase the expression of pyruvate dehydrogenase lipoamide kinase isozyme 4 (PDK4), a mitochondrial protein that regulates the tricarboxylic acid cycle (TCA) cycle. The inhibitor of PDK4 or its specific siRNA can attenuate BBP-induced cell proliferation and ATP generation, which suggested the essential roles of PDK4 in BBP-induced glycolysis and proliferation. Further, BBP can increase the mRNA stability of PDK4, while had no effect on its transcription and protein stability. miR-15b-5p can bind with the 3'UTR of PDK4 to decrease its mRNA stability, while BBP can decrease the expression of miR-15b-5p in AML cells. Collectively, our data showed that BBP can trigger the malignancy of AML cells via regulation of miR-15b-5p/PDK4 signals.
... Prior to evaluating the in vitro ROS generation from dual-triggered radical therapy, we determined E2 concentrations of three human cancer cell lines with high, mid, and low E2 concentrations. 52 Figure S4). We further found that the murine melanoma cell line B16F10 exhibits a very high E2 concentration of 124.25 G 8.78 pg/10 6 cells ( Figure S5). ...
Article
Checkpoint blockade immunotherapy (CBI) elicits durable therapeutic responses by blocking T cell inhibitory pathways of tumors with pre-infiltrated T cells and/or high mutational burden to activate antitumor immunity but is ineffective against poorly immunogenic tumors. Immunogenic radiotherapy, photodynamic therapy (PDT), and chemotherapy have thus been examined as immunomodulatory adjuvants to augment CBI. Dysregulated hormone production has long been linked to tumorigenesis and poor prognosis of various cancers. Herein, we report the use of a Cu-porphyrin nanoscale metal-organic framework (nMOF) to mediate synergistic hormone-triggered chemodynamic therapy (CDT) and light-triggered PDT. The combination of CDT/PDT-based radical therapy with a programmed cell-death ligand 1 blockade effectively extends the local therapeutic effects of CDT/PDT to distant tumors via abscopal effects on mouse tumor models with high levels of estradiol. Our work thus establishes the feasibility of combining nMOF-mediated radical therapy with CBI to elicit systemic antitumor immunity in hormonally dysregulated tumor phenotypes.
... This is in opposition to their mitogenic hydroxylated counterparts (2/4OH-CE) that can be metabolized into quinones leading to the formation of quinone adducts and oxidative DNA damage [17]. These reactive estrogen metabolites are critical in the initiation of breast and prostate cancers as well as non-Hodgkin lymphoma, and have been found in abundance in leukemia cell lines of the NCI-60 human tumor cell lines panel [44,45]. This may indicate that increased methylation of estrogens could be beneficial for CLL patients by preventing the formation and accumulation of damaging catechol estrogen metabolites and/or through their intrinsic beneficial effects. ...
Article
Estrogen receptor is an important target in breast cancer. Serotonin receptors (5‐HT 2A and 5‐HT 2C , in particular) were investigated for a potential role in development and progression of breast cancer. Ligands that interact with estrogenic receptors influence the emotional state of females. Thus, designing selective estrogen receptor modulator (SERM) analogs with potential serotonergic activity is a plausible approach. The dual ligands can augment cytotoxic effect of SERMs, help in both physical and emotional menopausal symptom relief, enhance cognitive function and support bone health. Herein, we report triarylethylene analogs as potential candidates for treatment of breast cancer. Compound 2e showed (ERα relative β‐ galactosidase activity = 0.70), 5‐HT 2A (K i = 0.97 µM), and 5‐HT 2C (K i = 3.86 µM). It was more potent on both MCF‐7 (GI 50 = 0.27 µM) and on MDA‐MB‐231 (GI 50 = 1.86 µM) compared to tamoxifen (TAM). Compound 4e showed 40 times higher antiproliferative activity on MCF‐7 and 15 times on MDA‐MBA compared to TAM. Compound 4e had higher average potency than TAM on all nine tested cell line panels. Our in‐silico model revealed the binding interactions of compounds 2 and 2e in the three receptors; further structural modifications are suggested to optimize binding to the ERα, 5‐HT 2A , and 5‐HT 2C .
Article
Acute myeloid leukemia (AML) is a cancer of hematopoietic stem cells with a rapid progression. The progression of AML can be regulated by estrogenic signals. Our present data showed that an industrial endocrine‐disrupting chemical, bisphenol A (BPA), can promote the proliferation of AML cells and decrease their sensitivity to daunorubicin and cytarabine treatment. Among the tested cytokines, BPA treatment can decrease the expression of interleukin‐4 (IL‐4) while increasing the expression of IL‐6. Overexpression of IL‐4 or neutralization antibody of IL‐6 (anti‐IL‐6) can attenuate BPA‐induced proliferation of AML cells and reverse BPA‐suppressed chemosensitivity. Furthermore, activation of nuclear factor kappa B is essential for BPA‐induced upregulation of IL‐6 in AML cells. As to IL‐4, BPA can increase the expression of NFAT1 to inhibit its transcription. Collectively, our data showed that BPA can trigger the malignancy of AML cells via regulation of IL‐4 and IL‐6.
Chapter
Cancer is considered as one of the deadliest diseases in the medical field. Finding novel and efficient natural compounds has been a major concern for cancer research and therapy. The abundant diversity of marine organisms, especially marine microorganisms, has expanded the sources of natural products for the screening of antitumor drugs. Thus, it is urgent to solve how to more efficiently screen marine microbial antitumor compounds. During marine microbe stress responses to virus infection, microorganisms must produce antiviral secondary metabolites to fight against bacteriophage infection. Due to the similarity of metabolic disorder for virus-infected microbes and tumorigenesis, the antiviral compounds possess antitumor capacity, which can restore the disordered metabolism of cancer cells to normal metabolic homeostasis.
Article
Full-text available
Leber's hereditary optic neuropathy, the most frequent mitochondrial disease due to mitochondrial DNA point mutations in complex I, is characterized by the selective degeneration of retinal ganglion cells, leading to optic atrophy and loss of central vision prevalently in young males. The current study investigated the reasons for the higher prevalence of Leber's hereditary optic neuropathy in males, exploring the potential compensatory effects of oestrogens on mutant cell metabolism. Control and Leber's hereditary optic neuropathy osteosarcoma-derived cybrids (11778/ND4, 3460/ND1 and 14484/ND6) were grown in glucose or glucose-free, galactose-supplemented medium. After having shown the nuclear and mitochondrial localization of oestrogen receptors in cybrids, experiments were carried out by adding 100 nM of 17β-oestradiol. In a set of experiments, cells were pre-incubated with the oestrogen receptor antagonist ICI 182780. Leber's hereditary optic neuropathy cybrids in galactose medium presented overproduction of reactive oxygen species, which led to decrease in mitochondrial membrane potential, increased apoptotic rate, loss of cell viability and hyper-fragmented mitochondrial morphology compared with control cybrids. Treatment with 17β-oestradiol significantly rescued these pathological features and led to the activation of the antioxidant enzyme superoxide dismutase 2. In addition, 17β-oestradiol induced a general activation of mitochondrial biogenesis and a small although significant improvement in energetic competence. All these effects were oestrogen receptor mediated. Finally, we showed that the oestrogen receptor β localizes to the mitochondrial network of human retinal ganglion cells. Our results strongly support a metabolic basis for the unexplained male prevalence in Leber's hereditary optic neuropathy and hold promises for a therapeutic use for oestrogen-like molecules.
Article
The mechanism of the antiproliferative activity of tamoxifen on melanoma cells in vitro and in vivo is poorly understood, as it is not mediated by the antiestrogenic properties of tamoxifen. Using a whole-cell assay and nuclear and cytosolic radio-binding experiments with [3H]-estradiol as tracer, we found that MNT1, M10, and M14 melanoma cell lines as well as primary tumors expressed type II estrogen binding sites that bind tamoxifen and the flavonoid quercetin with similar affinity (KD 10–25 nM). Cell count and clonogenic assay showed both compounds to inhibit melanoma cell growth in a concentration-dependent manner in the range of concentrations between 1 nM and 1 M. Neither the pure antiestrogen ICI-182780 nor the 3-rhamnosylglucoside of quercetin, rutin, bound to type II estrogen binding sites or inhibited cell growth. Our results suggesting that tamoxifen and quercetin can inhibit melanoma cell growth by interacting with type II estrogen binding sites help explain the reported effectiveness of tamoxifen, particularly in estrogen-receptor-negative tumors, and stress the potential role of quercetin in the treatment of melanoma.Keywords: antiestrogens, antitumor agent, estrogen receptor, flavonoids
Chapter
Multiple lines of evidence support a central role of hormones in the etiology of breast, endometrial and ovarian cancers. Evidence of an association between circulating hormones and these cancers varies by both hormone and cancer site, with the most consistent associations observed for sex steroid hormones and breast cancer risk among postmenopausal women. Recently, evidence has begun to accumulate suggesting an important role for endogenous hormones in premenopausal breast cancer, endometrial cancer and possibly ovarian cancer. In this chapter, prospective epidemiologic studies, where endogenous hormones are measured in study subjects prior to disease diagnosis, are summarized. Overall, a strong positive association between breast cancer risk and circulating levels of both estrogens and testosterone has now been well confirmed among postmenopausal women; women with hormone levels in the top 20% of the distribution (versus bottom 20%) have a two-to-three-fold higher risk of breast cancer. Evidence among premenopausal women is more limited, though increased risk associated with higher levels of testosterone is consistent. Evidence to date of hormonal associations for endometrial cancer is limited, though a strong association with sex steroid hormones is suggested. Studies of ovarian cancer have been few and small with no consistent associations observed with endogenous hormones. Clearly more evaluation is needed to confirm the role of endogenous hormones in premenopausal breast cancer, endometrial cancer and ovarian cancer.
Article
Many studies have reported a correlation between elevated estrogen blood levels and breast cancer and this observation has raised controversy concerning the long-term use of hormonal replacement therapy. This review will not address further this controversial topic; but rather, this review focuses on the role of estrogen signaling in first, the normal development of the breast and second, how alterations of this signaling pathway contribute to breast cancer.
Article
Aging of the skin is associated with skin thinning, atrophy, dryness, wrinkling, and delayed wound healing. These undesirable aging effects are exacerbated by declining estrogen levels in postmenopausal women. With the rise in interest in long-term postmenopausal skin management, studies on the restorative benefits that estrogen may have on aged skin have expanded. Systemic estrogen replacement therapy (ERT) has been shown to improve some aspects of skin. Estrogen restores skin thickness by increasing collagen synthesis while limiting excessive collagen degradation. Wrinkling is improved following estrogen treatment since estrogen enhances the morphology and synthesis of elastic fibers, collagen type III, and hyaluronic acids. Dryness is also alleviated through increased water-holding capacity, increased sebum production, and improved barrier function of the skin. Furthermore, estrogen modulates local inflammation, granulation, re-epithelialization, and possibly wound contraction, which collectively accelerates wound healing at the expense of forming lower quality scars. Despite its promises, long-term ERT has been associated with harmful systemic effects. In the search for safe and effective alternatives with more focused effects on the skin, topical estrogens, phytoestrogens, and tissue-specific drugs called selective estrogen receptor modulators (SERMs) have been explored. We discuss the promises and challenges of utilizing topical estrogens, SERMs, and phytoestrogens in postmenopausal skin management.
Article
Multiple lines of evidence support a central role of hormones in the etiology of breast, endometrial and ovarian cancers. Evidence of an association between circulating hormones and these cancers varies by both hormone and cancer site, with the most consistent associations observed for sex steroid hormones and breast cancer risk among postmenopausal women. Recently, evidence has begun to accumulate suggesting an important role for endogenous hormones in premenopausal breast cancer, endometrial cancer and possibly ovarian cancer. In this chapter, prospective epidemiologic studies, where endogenous hormones are measured in study subjects prior to disease diagnosis, are summarized. Overall, a strong positive association between breast cancer risk and circulating levels of both estrogens and testosterone has now been well confirmed among postmenopausal women; women with hormone levels in the top 20% of the distribution (versus bottom 20%) have a two-to-three-fold higher risk of breast cancer. Evidence amongpremenopausal women is more limited, though increased risk associated with higher levels of testosterone is consistent. Evidence to date of hormonal associations for endometrial cancer is limited, though a strong association with sex steroid hormones is suggested. Studies of ovarian cancer have been few and small with no consistent associations observed with endogenous hormones. Clearly more evaluation is needed to confirm the role of endogenous hormones in premenopausal breast cancer, endometrial cancer and ovarian cancer.
Article
Lung cancer is the leading cause of cancer mortality in both women and men worldwide but gender differences exist in their clinical and biological manifestations. In particular, among life time non-smoker, female are far more likely to develop lung carcinoma than male. Recent studies demonstrated that estrogens are synthesized in situ in both male and female lung cancers through aromatase, suggesting that sex steroid may contribute to the pathogenesis and development of lung carcinoma. In addition, human lung carcinomas have been recently demonstrated to be frequently associated with expression of estrogen receptors in both male and female patients and a lower expression of aromatase was reported to be associated with better prognosis. Preclinical studies further demonstrated that aromatase inhibitor (AI) suppressed the lung tumor growth both in vitro and in vivo. These findings all suggest a potential role of intratumoral aromatase in biological behavior of non-small cell lung cancer (NSCLC), the most common form of human lung malignancy. Therefore, AIs may become viable therapeutic options for disease management in NSCLC patients but further studies are definitely required to obtain a better understanding of the potential roles of intratumoral aromatase expression as a predictive biomarker for clinical outcome in these NSCLC patients.
Article
Estrogen plays a critical role in female reproduction but has also been reported to have important roles in various target tissues expressing estrogen receptor (ER) α and/or ERβ in both male and female. ERs especially ERβ have been demonstrated to be present and functional in both normal human lung and its disorders including cancer. Non-small cell lung carcinomas (NSCLCs) are well-known to be composed of heterogeneous groups. Squamous cell carcinoma is the most common subtype in men, but adenocarcinoma is the most common histologic subtype in women. Therefore, sex steroid hormones such as estrogens have been considered to play some roles in NSCLC. In particular, results of several epidemiological analyses pointed out the association between physiological or artificial alterations of hormone status such as menstruation and postmenopausal administration of hormone replacement therapy and lung cancer risks or its development especially in female subjects. In NSCLC tissues, intratumoral estrogen synthesis via aromatase, which is a key enzyme in the estrogen synthesis involved in aromatization of androgens into estrogens, has recently become of clinical interest as a possible target of therapy. Therefore, in this review, we focused on the potential of an endocrine therapy in NSCLC using clinically available inhibitors of estrogen and aromatase actions.
Article
Skin is a non classical target tissue for estrogens, whose biological and mechanical properties are affected by the hormonal deprivation occurring at the menopause. Estrogen-related receptors (ERR), closely related to the estrogen receptors (ER), constitute a subfamily of orphan receptors, interfering with ER-mediated signalling pathways. The expression of ERRβ has been detected in only a few adult tissues so far, such as the prostate and the inner ear. Here, we demonstrate the expression of ERRβ in normal human skin. ERRβ was detected in human epidermis, in both keratinocytes and Langerhans cells, by immunohistochemistry. These results were validated on freshly extracted epidermal cells and on monolayer-cultured keratinocytes by RT-PCR and western blotting (WB), suggesting the implication of ERRβ in skin immunity and endocrine effects.
Article
Estrogens produced as a result of intratumoral aromatization has been recently shown to play important roles in proliferation of human non-small cell lung carcinomas (NSCLC), but the details have remained largely unknown. Therefore, in this study, we evaluated the possible roles of intratumoral aromatase in NSCLCs as follows: (a) evaluation of intratumoral localization of aromatase mRNA/protein in six lung adenocarcinoma cases using laser capture microdissection combined with quantitative reverse transcriptase-PCR and immunohistochemistry; (b) examination of the possible effects of isolated stromal cells from lung carcinoma tissues on aromatase mRNA transcript expression in lung carcinoma cell lines (A549 and LK87) through a coculture system; and (c) screening of cytokines derived from stromal LK001S and LK002S cells using cytokine antibody arrays and subsequent evaluation of effects of these cytokines on aromatase expression in A549 and LK87. Both aromatase mRNA and protein were mainly detected in intratumoral carcinoma cells but not in stromal cells. Aromatase expression of A549 and LK87 was upregulated in the presence of LK001S or LK002S cells. Several cytokines such as interleukin-6 (IL-6), oncostatin M, and tumor necrosis factor-alpha, all known as inducible factors of aromatase gene, were detected in conditioned media of LK001S and LK002S cells. Treatment of both oncostatin M and IL-6 induced aromatase gene expression in A549 an LK87, respectively. These results all indicated that intratumoral microenvironments, especially carcinoma-stromal cell interactions, play a pivotal role in the regulation of intratumoral estrogen synthesis through aromatase expression in human lung adenocarcinomas.