ArticlePDF Available

Importance of dosage standardization for interpreting transcriptomal signature profiles: Evidence from studies of xenoestrogens

Authors:

Abstract and Figures

To obtain insights into similarities and differences in the biological actions of related drugs or toxic agents, their transcriptomal signature profiles (TSPs) have been examined in a large number of studies. However, many such reports did not provide proper justification for the dosage criteria of each agent. Using a well characterized cell culture model of estrogen-dependent proliferation of MCF7 human breast cancer cells, we demonstrate how different approaches to dosage standardization exert critical influences on TSPs, leading to different and even conflicting conclusions. Using quantitative cellular response (QCR)-based dosage criteria, TSPs were determined by Affymetrix microarray when cells were proliferating at comparable rates in the presence of various estrogens. We observed that TSPs of the xenoestrogens (e.g., genistein or bisphenol A) were clearly different from the TSP of 17β-estradiol; namely, the former strongly enhanced expression of genes involved in mitochondrial oxidative phosphorylation, whereas the latter showed minimal effects. In contrast, TSPs for genistein and 17β-estradiol were indistinguishable by using the marker gene expression-based dosage criteria, conditions in which there was comparable expression of the mRNA transcripts for the estrogen-inducible WISP2 gene. Our findings indicate that determination and interpretation of TSPs in pharmacogenomic and toxicogenomic studies that examine the transcriptomal actions of related agents by microarray require a clear rationale for the dosage standardization method to be used. We suggest that future studies involving TSP analyses use quantitative and objective dosage standardization methods, such as those with quantitative cellular response or marker gene expression-based dosage criteria. • breast cancer • estrogen • pharmacogenomics • toxicogenomics • transcriptome
Content may be subject to copyright.
Importance of dosage standardization for interpreting
transcriptomal signature profiles: Evidence from
studies of xenoestrogens
Toshi Shioda*
, Jessica Chesnes*, Kathryn R. Coser*, Lihua Zou
, Jingyung Hur*, Kathleen L. Dean*,
Carlos Sonnenschein
§
, Ana M. Soto
§
, and Kurt J. Isselbacher*
*Department of Tumor Biology and Molecular Profiling Laboratory, Massachusetts General Hospital Center for Cancer Research, Charlestown, MA 02129;
Division of Computational Biology, Harvard Bauer Center for Genomics Research, Cambridge, MA 02138; and
§
Department of Anatomy and Cell Biology,
Tufts University School of Medicine, Boston, MA 02111
Contributed by Kurt J. Isselbacher, June 26, 2006
To obtain insights into similarities and differences in the biological
actions of related drugs or toxic agents, their transcriptomal
signature profiles (TSPs) have been examined in a large number of
studies. However, many such reports did not provide proper
justification for the dosage criteria of each agent. Using a well
characterized cell culture model of estrogen-dependent prolifera-
tion of MCF7 human breast cancer cells, we demonstrate how
different approaches to dosage standardization exert critical in-
fluences on TSPs, leading to different and even conflicting conclu-
sions. Using quantitative cellular response (QCR)-based dosage
criteria, TSPs were determined by Affymetrix microarray when cells
were proliferating at comparable rates in the presence of various
estrogens. We observed that TSPs of the xenoestrogens (e.g.,
genistein or bisphenol A) were clearly different from the TSP of
17
-estradiol; namely, the former strongly enhanced expression of
genes involved in mitochondrial oxidative phosphorylation,
whereas the latter showed minimal effects. In contrast, TSPs for
genistein and 17
-estradiol were indistinguishable by using the
marker gene expression-based dosage criteria, conditions in which
there was comparable expression of the mRNA transcripts for the
estrogen-inducible WISP2 gene. Our findings indicate that deter-
mination and interpretation of TSPs in pharmacogenomic and
toxicogenomic studies that examine the transcriptomal actions of
related agents by microarray require a clear rationale for the
dosage standardization method to be used. We suggest that future
studies involving TSP analyses use quantitative and objective
dosage standardization methods, such as those with quantitative
cellular response or marker gene expression-based dosage criteria.
breast cancer estrogen pharmacogenomics toxicogenomics
transcriptome
M
icroarray determinations of transcriptomal changes in-
duced by biologically active substances have bec ome in-
creasingly popular in pharmacogenomics research. For example,
a large number of studies have reported time- and dose-
dependent aspects of transcriptomal changes induced by estro-
gen ic and antiestrogenic agents in cell culture and an imal models
(1–17). To obtain insights into similarities and differences in the
ef fects of multiple hor monal agents, it has been common
practice to determine their transcriptomal signature profiles
(TSPs; ref. 18). TSP analyses of antiestrogens such as tamoxifen,
raloxifen, or fulvestrant have suggested a molecular basis for
their partial estrogen-like activities (1–6). Studies of TSPs for
17
-estradiol (E
2
) and xenoestrogens have provided insights into
a genomics-based classification of estrogens and their mecha-
n isms of action (6–9).
To determine TSPs of hormonally active agents, estrogen
t arget cells and tissues such as estrogen-dependent human
breast cancer cell cultures (1–7, 12–14) and the rodent uterus
(8, 9, 16, 17) have been used as standards in in vitro and in vivo
model systems, respectively. For both systems, the time-
dependent aspects of the agent-induced transcriptomal
changes have been well characterized (3, 5, 9, 12–14, 16, 17).
In contrast, relatively few studies have systematically examined
the dosage-dependent aspects of the hor mone-induced tran-
scriptomal changes (6, 19). In many studies, TSPs of estrogens
and antiestrogens were deter mined by using a relatively high
single dose of each agent. For example, whereas the prolifer-
ative effect of E
2
on estrogen-t arget MCF7 human breast
cancer cells is saturated at 0.1 nM (12, 19), w ith few
exceptions (12, 19) most studies have determined TSPs of this
model using E
2
at 1–10 nM (1–7, 10, 13, 14). However, we
observed that the transcriptomal ef fects of E
2
on MCF7 cells
showed a strong dosage dependency, and that high concen-
trations of E
2
(0.1 nM) induced ex pression of mRNA
transcripts for TGF-
and stromal cell-derived factor-1 (SDF-
1), which c ould not be induced by lower c oncentrations of E
2
,
yet these lower concentrations induced maximal cell prolifer-
ation (19). Thus, interpret ation of TSP dat a using a single
saturating c oncentration of a hor monal agent may have sig-
n ificant limit ations.
In the present study, by characterizing the transcriptomal
ef fects of E
2
and xenoestrogens in MCF7 cells, we demonstrate
the importance of using objective and quantitative criteria to
st andardize the dosage of hormonal agents in TSP-based phar-
mac ogenomics. We propose two approaches to dosage st andard-
ization and discuss their advantages and limitations.
Results
To characterize the dose-dependent aspects of estrogen effects
on MCF7 cell proliferation, we measured the increase in cell
yield during 120 h of exposure to varying c oncentrations of E
2
and of xenoestrogens (Fig. 1A). E
2
, the natural an imal estrogen,
strongly enhanced MCF7 cell proliferation at 3–100 pM.
Bisphenol A and p-nonylphenol, representative plastic-related
xenoestrogens, enhanced MCF7 cell proliferation at 3–100 nM.
Gen istein and daidzein, representative isoflavone phytoestro-
gens that bind preferably to estrogen receptor (ER)
(20), were
weak xenoestrogens that stimulated MCF7 cell proliferation at
c oncentrations of 0.1
M. Tris (4-hydroxyphenyl)-4-propyl-1-
pyrazole (PPT), a synthetic selective agonist for ER
, increased
Conflict of interest: no conflicts declared.
Freely available online through the PNAS open access option.
Abbreviations: E
2
,17
-estradiol; MGE, marker gene expression; PAR, pairwise angle ratio;
PPT, 1,3,5-tris(4-hydroxyphenyl)-4-propyl-1-pyrazole; QCR, quantitative cellular response;
TSP, transcriptomal signature profile.
Data deposition: The microarray data presented in this study have been deposited in the
Gene Expression Omnibus (GEO) database, www.ncbi.nlm.nih.govgeo (accession no.
GSE5200).
To whom correspondence may be addressed. E-mail: tshioda@partners.org or kisselbacher@
partners.org.
© 2006 by The National Academy of Sciences of the USA
www.pnas.orgcgidoi10.1073pnas.0605341103 PNAS
August 8, 2006
vol. 103
no. 32
12033–12038
GENETICS
MCF7 cell proliferation relatively strongly at 0.1–10 nM. Our
120-h assay clearly demonstrated a quantitative dif ference be-
t ween 30 pM E
2
and 3
M genistein on MCF7 cell proliferation
ef fects. However, there was no observed difference in the ef fects
of 30 pM E
2
and 10
M genistein. Assays of cell proliferation at
shorter time periods of estrogen exposure (e.g., 48 h) were not
quantit atively dif ferent in distinguishing the effects of 30 pM E
2
and 3
M genistein (data not shown).
Our preliminary ex periments on the time-dependent aspects
of the transcriptomal effects of E
2
in MCF7 cells revealed that
many known estrogen-inducible genes reached their maximum
levels of expression only after 48 h of exposure. A previous study
by us showed consistent reproducibility of the transcriptomal
profiling of MCF7 cells when exposed to varying concentrations
of E
2
for 48 h (19). However, MCF7 cell transcriptomes deter-
mined after 120 h of exposure to E
2
showed poorer reproduc-
ibilit y, presumably reflecting that cells were no longer synchro-
n ized in their transcriptomal responses to E
2
(dat a not shown).
Therefore, although the estrogen effects on cell proliferation
were assayed after 120 h of exposure, transcriptomal effects were
deter mined at 48 h.
To determine TSPs of xenoe strogens using DNA microarrays, we
defined two criteria for dosage standardization. The first, quanti-
tative cellular response (QCR)-based dosage criteria, identified
doses that induce an equal QCR. Using the QCR criteria, estrogen
concentrations that supported MCF7 cell proliferation with a
strength equal to 30 pM E
2
were identified, namely, 3 nM PPT, 10
nM bisphenol A, 10 nM p-nonylphenol, 1
M daidzein, and 10
M
genistein (Fig. 1 A, red circles). Interestingly, even though 30 pM E
2
and 10
M genistein equally enhanced MCF7 cell proliferation, the
latter induced expre ssion of the mRNA transcripts for WISP2
3-fold more strongly than the former (Fig. 4B, which is published
as supporting information on the PNAS web site). Because WISP2
is a representative E
2
-inducible gene whose expression is strongly
enhanced from 1 to 60 pM E
2
in MCF7 cells (19), our observation
demonstrated a significant discrepancy between the proliferative
and the transcriptional effects of estrogens. By exposing MCF7 cells
to varying concentrations of genistein, we observed that 30 pM E
2
and 3
M genistein equally induced WISP2 mRNA (48 h of
exposure; Fig. 1B). However, at this concentration, genistein stim-
ulated MCF7 cell proliferation only half as much as 30 pM E
2
(120
h of exposure; Fig. 1 A). Based on these observations, we defined
the second criteria of estrogen dosage standardization, namely,
marker gene expression (MGE)-based dosage criteria that identi-
fied concentrations of E
2
(30 pM) and genistein (3
M) that equally
induced mRNA for WISP2 (Fig. 1A, blue circles).
The xenoestrogen TSPs were determined by using Af fymetrix
(Sant a Clara, CA) GeneChip DNA microarrays by exposing
MCF7 cells to these agents for 48 h at the dosage chosen by the
QCR and MGE criteria. Initial characterization of the xe-
noestrogen TSPs with 20 represent ative estrogen-inducible
genes (19) revealed that the transcriptomal effects of E
2
and
gen istein at the dosage deter mined by the MGE criteria were
remark ably similar (Fig. 4A). In c ontrast, transcriptomal effects
of E
2
and the xenoestrogens (including genistein) on these
representative marker genes were clearly different when their
dosage was determined by the QCR criteria (Fig. 4 B–D). The
ex pression profiles of these 20 marker genes were similar be-
t ween genistein and daidzein (Fig. 4B ). Profiles for bisphenol A
and p-nonylphenol also showed apparent similarit y (Fig. 4C).
The profile for PPT fit between the phytoestrogen pattern and
the plastic estrogen pattern (Fig. 4D). The remarkable differ-
ences observed among the TSPs using the QCR criteria sug-
gested that different estrogens have discrete transcriptomal
actions that can be used to discriminate the E
2
, soybean isofla-
vones, and man-made chemical xenoestrogens even when their
phar macological or toxicological effects are comparable. In
c ontrast, the TSPs of E
2
and genistein, which were clearly
dif ferent using the QCR criteria, were strikingly similar using the
MGE criteria, suggesting that the transcriptomal actions of E
2
and genistein were essentially identical. These conflicting ob-
servations exemplify the significant but often-overlooked risk of
interpreting the transcriptomal profiling data beyond the limi-
t ations of the experimental conditions.
The TSPs of estrogens were further characterized by hierar-
chical clustering analysis involving 1,675 informative genes
whose expression was strongly affected by at least one of the
estrogens (Fig. 2A). This analysis involved 24 TSP sets repre-
senting seven dif ferent estrogens and control vehicle. The TSPs
of each agent were determined by three independently per-
for med cell culture experiments and were grouped into five
major classes. Consistent with the in itial characterization involv-
ing the 20 selected genes, E
2
and gen istein at the concentrations
deter mined by MGE criteria [shown as genistein(L) in Fig. 2 A]
for med a single class (TSP names shown in red). The t wo
phy toestrogens (gen istein and daidzein) formed a separate class
(TSP names in blue), and the t wo plastic-related chemicals
(bisphenol A and p-nonylphenol) formed another class (TSP
names in green). PPT and vehicle (0.1% ethanol) showed
patterns different from those observed for the above-mentioned
estrogens (TSP names in black or purple, respectively).
A
B
Fig. 1. Determination of xenoestrogen concentrations based on QCR and
MGE-based dosage criteria. (A) Estrogen-dependent proliferation of MCF7
cells. Effects of xenoestrogens on cell proliferation were determined by the
120-h E-SCREEN assay, as described (19). The number of cells in the wells
supplemented with 100 pM E
2
was determined by a Coulter (Fullerton, CA) cell
counter and defined as 100% relative cell number, which corresponded to
50% confluence. Numbers of cells cultured in the presence of the xenoestro-
gens were expressed relative to this condition. Each point represents mean
SEM (n 5). Xenoestrogen concentrations determined by QCR criteria are
indicated by red circles; E
2
and genistein concentrations determined by MGE
criteria are indicated by blue circles. (B) Effects of 48 h of exposure to E
2
(30 pM)
and genistein (3
M) on expression of the WISP2 mRNA transcripts in MCF7
cells. Real-time quantitative PCR determination of amount of WISP2 mRNA by
three independent sets of experiments is shown.
12034
www.pnas.orgcgidoi10.1073pnas.0605341103 Shioda et al.
Fig. 2. Clustering analysis of xenoestrogen effects on MCF7 cell transcriptome. (A) Heat-map representation of 2D hierarchical clustering analysis of the TSPs
of xenoestrogens. Concentrations of the estrogenic agents were determined with QCR criteria except genistein(L) (low concentration), which was determined
by using MGE criteria. Bars below the heat map indicate seven subclusters of genes. (B) Statistical significance of similarities between the TSPs of xenoestrogens.
Histogram shows the joint null distribution of the PAR
ij
. Dots indicate observed PAR
ij
between the indicated pairs of groups of xenoestrogen TSPs. The P values
indicate how unlikely the observed TSP similarity would be to happen by chance. (C) Details of xenoestrogen effects on expression of representative
estrogen-discriminating marker genes. Each column represents a marker gene, and the points indicate fold changes in mRNA expression over vehicle control in
the log scale. The pattern of the representative unchanged genes is also shown as control. BPA, bisphenol A; PNP, p-nonylphenol.
Shioda et al. PNAS
August 8, 2006
vol. 103
no. 32
12035
GENETICS
To objectively examine the similarities between the TSP classes,
pairwise angle ratios (PARs) were calculated as measurements for
distance s between two TSP groups. To obtain the P values for the
TSP similarities, the null distribution of PARs was calculated by
using randomly generated PARs, and the locations of the measured
PARs were determined (Fig. 2B). As expected from the heat-map
pattern (Fig. 2 A), a strong similarity between TSPs for E
2
and the
low-concentration genistein (3
M; MGE criteria) was confirmed
by this approach (P 0.0002). A strong similarity between TSPs for
daidzein and high-concentration genistein (10
M, QCR criteria)
was also shown (P 0.0002). A marginally significant similarity
between TSPs for bisphenol A and p-nonylphenol was demon-
strated by the PAR location at the foot of the null distribution curve
(P 0.002).
The clustering analysis identified seven subclusters of class-
discriminating marker genes (Fig. 2 A, bottom bars, and Table 1,
which is published as supporting information on the PNAS web
site). Subclusters 1a, 1b, and 1c were similar in their expression
patterns, together forming cluster 1; subclusters 3a and 3b formed
cluster 3; and subclusters 2 and 4 were unique. Expression profiles
of 20 genes representing each subcluster are shown in Fig. 2C
(subclusters 1a, 2, 3a, and 4) and Fig. 5, which is published as
supporting information on the PNAS web site (all subclusters).
Profiles of unchanged genes are also shown as control. Cluster 1
consisted of genes induced by the xenoestrogens but not by E
2
. The
low MGE-criteria concentration of genistein [3
M, shown as
genistein(L)] also did not induce the cluster 1 genes. Cluster 2
involved genes strongly down-regulated by E
2
as well as by low and
high (10
M, QCR criteria) concentrations of genistein and daid-
zein. Expre ssion of these cluster 2 genes was only modestly affected
by bisphenol A, p-nonylphenol, or PPT. In contrast, expression of
cluster 3 genes was strongly up-regulated by E
2
, by low and high
concentrations of genistein and daidzein, but not by bisphenol A or
PPT. Interestingly, the cluster 3 genes were induced by p-
nonylphenol, demonstrating the differential effects of the two
plastic-related xenoestrogens. Cluster 4 involved genes most
strongly induced by daidzein, high-concentration genistein, and
p-nonylphenol; they were induced relatively weakly by E
2
, low-
concentration genistein, and bisphenol A. Taken together, these
results provide further support for the following concepts: (i) TSPs
determined by using the QCR-criteria dosage reflect the divergent
transcriptomal effects of the estrogenic agents when their cell
proliferation responses are set to be equal; and (ii) the TSPs
determined by using the MGE-criteria dosage reflect the maximum
degree of similarity in their transcriptomal effects.
To predict the biological outcomes of the differential transcrip-
tomal effects of the xenoestrogens, we performed gene ontology
pathway analyses using the Kyoto Encyclopedia of Genes and
Genomes (KEGG) PATHWAY database (21). As expected from
their cell proliferation effects, large numbers of genes involved in
cell cycle progression were up-regulated by both E
2
and the
xenoestrogens (Table 2, which is published as supporting informa-
tion on the PNAS web site). Genes involved in purine and pyrim-
idine metabolism that are required for DNA synthesis as well as
genes involved in steroid biosynthesis were also significantly acti-
vated by the e strogens. Interestingly, of the 103 gene s involved in
mitochondrial oxidative phosphorylation, 4683% were up-
regulated by the xenoestrogens. Fig. 6, which is published as
supporting information on the PNAS web site, shows that the
p-nonylphenol up-regulated genes were involved in all five oxidative
phosphorylation c omplexes. However, E
2
, PPT, and low-
concentration genistein did not significantly affect the expre ssion of
these genes. Analyses using the Gene Ontology database (22)
revealed re sults similar to the KEGG database (data not shown).
To compare the effects of each xenoestrogen on the expression
of oxidative phosphorylation-related genes, amounts of these genes
(82 genes, p-nonylphenol-induced) were plotted in Fig. 3A.Al-
though expre ssion of most of these genes was not significantly
enhanced by E
2
or by low-concentration genistein, they were
increased 2-fold or greater by high-concentration genistein and
other xenoestrogens. PPT also significantly enhanced expression of
these genes but more weakly than other xenoestrogens. Expre ssion
of the mRNA transcripts for the uncoupling proteins (UCP-1, -2,
and -3) that reflect mitochondrial biogenesis (23) was not affected
by E
2
or by low-concentration genistein; interestingly, UCP-1
expre ssion was moderately enhanced by genistein (Fig. 3B). In
contrast, expression of the UCP genes was strongly enhanced by the
xenoestrogens, but expre ssion of GAPDH and
-actin (ACTB) was
unaffected by any estrogen. Thus, when the QCR criteria were used
for dosage standardization, the xenoestrogens (including genistein)
significantly increased expre ssion of the oxidative phosphorylation-
related genes, but E
2
did not. In contrast, when the MGE criteria
were used to determine concentrations of E
2
and genistein, neither
affected expression of these genes. These findings indicate that the
use of different criteria for dosage standardization for pharmacog-
enomics studie s may lead to different conclusions.
Fig. 3. Xenoestrogen induction of genes involved in mitochondrial oxidative
phosphorylation. (A) Xenoestrogen induction of 82 genes involved in oxida-
tive phosphorylation. The low [genistein(L), 3
M] and high concentrations
[genistein, 10
M] of genistein were determined by using MGE and QCR
criteria, respectively. Each point indicates fold increase in mRNA amounts in
estrogen-exposed MCF7 cells over vehicle-exposed control (average of three
microarray data, log scale). Averages of fold increase of all points for each
estrogen are shown on the top of the graph (mean SEM, n 82). (B)
Estrogen-induced expression of mRNA for mitochondrial uncoupling protein
(UCP1–3) and two housekeeping genes, GAPDH and
-actin (ACTB). Each bar
represents fold increase in mRNA amounts in estrogen-exposed MCF7 cells
over vehicle-exposed control (average of three microarray data).
12036
www.pnas.orgcgidoi10.1073pnas.0605341103 Shioda et al.
Discussion
Microarray technology is increasingly being used in pharmacolog-
ical and toxicological studies. Several guidelines such as Minimum
Information About a Microarray Experiment (MIAME) and its
toxicology-adapted version MIAMETox (24), as well as the
Minimum Information Needed for a Toxicology experiment (MIN-
Tox; ref. 25), have been proposed for standardized description of
experiments for pharmaco- and toxicogenomic studies. Unfortu-
nately, however, these guidelines do not specify any method for
dosage standardization in studying biologically active agents. Al-
though we have emphasized the critical importance of the dosage-
dependent aspects of the experimental determination of TSPs by
DNA microarray (19), this concern is often overlooked. For exam-
ple, microarray experiments comparing the TSPs of estrogen-
related agents are often performed with doses selected either
arbitrarily or empirically, namely, without apparent objective or
quantitative criteria for dosage standardization (1–7, 10, 13, 14).
To provide some guidance for dosage standardization of phar-
maco- and toxicogenomic experiments, in the present study, we
describe two approaches: the QCR criteria, that provide the TSPs
representing different agents known to induce an equal QCR; and
the MGE criteria, that provide the TSPs including reference marker
genes that are expre ssed equally in the presence of different agents
(Fig. 1). The TSPs determined using these two criteria were
remarkably different (Fig. 2) and thus led to different and con-
flicting conclusions (Fig. 3). Experiments using the QCR criteria
indicate that, in MCF7 cells, the TSPs of xenoestrogens differed
significantly from the TSP of E
2
(Fig. 2). TSP pathway analysis
showed that the xenoestrogens (including genistein) significantly
increased expre ssion of the genes involved in mitochondrial oxida-
tive phosphorylation, whereas E
2
showed no effects on expre ssion
of these genes (Fig. 3). However, it must be noted that these
observations were valid only at estrogen concentrations that en-
hanced MCF7 cell proliferation to comparable levels (Fig. 1). In
contrast, experiments performed using the MGE criteria convinc-
ingly demonstrated that the transcriptomal effects of genistein and
E
2
were practically identical (Fig. 2). In other words, the transcrip-
tomal effects of genistein and E
2
in MCF7 cells became indistin-
guishable at certain concentrations. Thus, the MGE criteria may be
more appropriate than the QCR criteria when the objective of a
study is to determine the maximum TSP similarities of different
agents. In contrast, the QCR criteria may be useful to characterize
difference s in mechanisms of action as well as side effects of agents
showing quantitatively similar biological actions. Table 3, which is
published as supporting information on the PNAS web site, sum-
marizes the features of different dosage criteria for the determi-
nation of TSPs.
The concept of the QCR and MGE dosage standardization
criteria should be applicable not only to studies of estrogenic
agents but more generally for other types of phar maco
toxic ogenomic studies involving both in vitro and in vivo models.
It is important to mention that the QCR and MGE criteria may
be applicable to studies involv ing human subjects or wildlife
an imals as long as a specific and quantitative agent-induced
phenot ype or marker gene is available (see Table 3). For
example, applying the MGE criteria, a clinical study may be able
to select patient-derived specimens whose drug-inducible MGEs
are comparable and ask whether the behavior of other genes is
similar when different drugs are used. Similarly, using the QCR
criteria, wildlife animals exposed to a group of environmental
c ontaminants showing similar and specific effects on a common
t arget tissue may be stratified before transcriptomal profiling. To
facilit ate dat abase-based bioinfor matics research on genomics
dat a, it would be important to have a description of the dosage
st andardization criteria (e.g., QCR, MGE, or empirical) when
TSPs and information on the doses of agents are incorporated
into databases. For this purpose, it may be helpful to extend the
MI AMETox (Minimum Information About a Microarray Ex-
perimentTox) and MIN-Tox (Minimum Information Needed
for a Toxicolog y Ex periment) guidelines accordingly.
In summary, we have pre sented evidence that different criteria
for dosage standardization of biologically active agents may result
in conflicting andor misleading conclusions of a pharmaco
toxicogenomic study. We have also called attention to the risk of
interpreting TSP data where hormonal doses were selected on
either an empirical or an arbitrary basis. We have also described
applications of two dosage-standardizing criteria, namely, QCR and
MGE criteria. To correctly interpret genomics studies generating
TSPs, it is critical to specify the methods and criteria used for dosage
standardization and to provide the rationale for their use.
Materials and Methods
Cell Culture. Culture c onditions and the 120-h E-SCREEN pro-
liferation assay of MCF7 cells (BUS stock) were previously
described (19). To deter mine the transcriptomal effects of
estrogens, cells were washed three times with phenol red-free
DMEM and incubated at 37°C fo r1hinthefinal wash. The
medium was then changed to phenol red-free DMEM supple-
mented with 5% charcoaldextran-stripped FCS (HyClone, Lo-
gan, UT), and cells were cultured for 48 h in the presence of
estrogens. All plasticware was carefully selected to minimize
xenoestrogen contamination. As reported (26), contamination
of labware-derived xenoestrogens exerted critical effects on both
MCF7 cell proliferation and gene expression.
Microarray Experiment and Data Analysis. Total RNA extraction and
Affymetrix U133A GeneChip DNA microarray experiments were
performed as described in our previous study (19). Twenty-four
microarray data sets, which represented eight different estrogen
treatments, were obtained from three independently performed
cell culture experiments for each treatment. To perform 2D hier-
archical clustering analysis, 22,283 genes on the array were filtered
by using the following criteria: (i) the scaled intensity value was
600 for at least three microarray data sets; (ii) the difference
between the maximum and minimum intensity value was 300; and
(iii) the standard deviation of the gene vector was 300. Expression
intensities of 1,675 informative genes were log-transformed and
analyzed by using Cluster software and visualized with TreeView
software (27). Complete linkage clustering analysis was performed
with median center for both genes and array data sets using
centered correlation as a similarity metric. Gene ontology analysis
was performed by using the GeneSifter.net on-line service (vizX-
labs, Seattle, WA), which served as a communication tool to the
Kyoto Encyclopedia of Genes and Genomes database (28) and the
Gene Ontology Consortium Database (22).
To objectively evaluate similarities of transcriptomal effects
bet ween t wo estrogens, we defined the PAR
ij
as
PAR
ij
cos
1
ij
min
k
i, j
l
i, j
cos
1
kl
1 i j 8, [1]
where
ij
is the sample correlation of gene expression under
treatments i and j (1 i j 8). The inverse cosine of the
sample correlation measures the angle between the two gene
ex pression vectors, with cos
1
(1) 0 when gene ex pression is
perfectly positively correlated under the two treatments and
cos
1
(1)
for perfect negative correlation. cos
1
(
ij
) can
therefore be interpreted as the gene-expression distance be-
t ween treatments i and j. When PAR
ij
, which is the ratio of these
dist ances, is significantly small, the transcriptomal effects of
treatments i and j are significantly close to each other. To assess
the significance of the observed PAR
ij
, the joint null distribution
of PAR
ij
was computed for a randomly sampled set of 1,675 genes
f rom the list of 22,283 genes. The observed PAR
ij
was c ompared
Shioda et al. PNAS
August 8, 2006
vol. 103
no. 32
12037
GENETICS
with the distribution of min
i,j
PAR
ij
across 5,000 samples from
this null distribution to obtain a P value as:
p
ij
f raction of simulated min
l,m
PAR
lm
smaller than observed PAR
ij
. [2]
Real-Time Quantitative PCR (RTQ-PCR). RTQ-PCR was performed
using TaqMan PCR Master Mix and ABI Prism 7700 thermal
c ycler (Applied Biosystems, Framingham, MA). PCR primers
for human WISP2 were 5-CATGAGAGGCACACCGAAGA
(sense) and 5- GCACCTTTGAGAGGAGGCAG (antisense),
and a TaqMan probe for this mRNA transcript was 5-VIC-
CCACCTCCTGGCCTTCTCCCTCC-TAMRA.
We thank Ben Wittner, Paul Grosu, and Reddy Gali for useful discus-
sions and Kremena Star for in itial contributions to microarray data
interpretation. We also thank James Signorovitch for his contributions
to the statistical evaluation of similarities between transcriptomal pro-
files. This study was funded in part by Susan Komen Breast Cancer
Foundation Grant BCTR0503620 (to T.S.), National Institutes of Health
R01 Grants CA82230 (to T.S.) and ES012301 (to A.M.S.), and the
Foundation for Research in Cell Biology and Cancer (T.S.).
1. Vendrell, J. A., Magnino, F., Danis, E., Duchesne, M. J., Pinloche, S., Pons, M.,
Birnbaum, D., Nguyen, C., Theillet, C. & Cohen, P. A. (2004) J. Mol.
Endocrinol. 32, 397–414.
2. Inoue, A., Yoshida, N., Omoto, Y., Oguchi, S., Yamori, T., Kiyama, R. &
Hayashi, S. (2002) J. Mol. Endocrinol. 29, 175–192.
3. Wang, D. Y., Fulthorpe, R., Liss, S. N. & Edwards, E. A. (2004) Mol.
Endocrinol. 18, 402–411.
4. Gadal, F., Starzec, A., Bozic, C., Pillot-Brochet, C., Malinge, S., Ozanne, V.,
Vicenzi, J., Buffat, L., Perret, G., Iris, F. & Crepin, M. (2005) J. Mol .
Endocrinol. 34, 61–75.
5. Frasor, J., Stossi, F., Danes, J. M., Komm, B., Ly ttle, C. R. & Katzenellenbogen,
B. S. (2004) Cancer Res. 64, 1522–1533.
6. Jorgensen, M., Vendelbo, B., Skakkebaek, N. E. & Leffers, H. (2000) Environ.
Health Perspect. 108, 403–412.
7. Terasaka, S., Aita, Y., Inoue, A., Hayashi, S., Nishigaki, M., Aoyagi, K., Sasak i,
H., Wada-Kiyama, Y., Sakuma, Y., Akaba, S., et al. (2004) Environ. Health
Perspect. 112, 773–781.
8. Watanabe, H., Suzuki, A., Goto, M., Lubahn, D. B., Handa, H. & Iguchi, T.
(2004) J. Mol. Endocrinol. 33, 243–252.
9. Moggs, J. G., Ashby, J., Tinwell, H., Lim, F. L., Moore, D. J., Kimber, I. &
Orphanides, G. (2004) Environ. Health Perspect. 112, 1137–1142.
10. Harvell, D. M., Richer, J. K., Allred, D. C., Sartorius, C. A. & Horwitz, K. B.
(2006) Endocrinology 147, 700–713.
11. Stossi, F., Barnett, D. H., Frasor, J., Komm, B., Lyttle, C. R. & Katzenellen-
bogen, B. S. (2004) Endocrinology 145, 3473–3486.
12. Lobenhofer, E. K., Bennett, L., Cable, P. L., Li, L., Bushel, P. R. & Afshari,
C. A. (2002) Mol. Endocrinol. 16, 1215–1229.
13. Soulez, M. & Parker, M. G. (2001) J. Mol. Endocr inol. 27, 259–274.
14. Frasor, J., Danes, J. M., Komm, B., Chang, K. C., Ly ttle, C. R. & Katzenel-
lenbogen, B. S. (2003) Endocrinology 144, 4562–4574.
15. Ma, X. J., Salunga, R., Tuggle, J. T., Gaudet, J., Enright, E., McQuary, P.,
Payette, T., Pistone, M., Stecker, K., Zhang, B. M., et al. (2003) Proc. Natl.
Acad. Sci . USA 100, 5974–5979.
16. Moggs, J. G., Tinwell, H., Spur way, T., Chang, H. S., Pate, I., Lim, F. L., Moore,
D. J., Soames, A., Stuckey, R., Currie, R., et al. (2004) Environ. Health Perspect.
112, 1589–1606.
17. Watanabe, H., Suzuki, A., Kobayashi, M., Takahashi, E., It amoto, M., Lubahn,
D. B., Handa, H. & Iguchi, T. (2003) J. Mol. Endocrinol. 30, 347–358.
18. Merrick, B. A. & Bruno, M. E. (2004) Curr. Opin. Mol. Ther. 6, 600607.
19. Coser, K. R., Chesnes, J., Hur, J., Ray, S., Isselbacher, K. J. & Shioda, T. (2003)
Proc. Natl . Acad . Sci . USA 100, 13994–13999.
20. Hall, J. M. & Korach, K. S. (2002) J. Biol. Chem. 277, 44455–44461.
21. Kanehisa, M., Goto, S., Kawashima, S., Okuno, Y. & Hattori, M. (2004) Nucleic
Acids Res. 32, D277–D280.
22. Ashburner, M., Ball, C. A., Blake, J. A., Botstein, D., Butler, H., Cherry, J. M.,
Dav is, A. P., Dolinski, K., Dwight, S. S., Eppig, J. T., et al . (2000) Nat. Genet.
25, 25–29.
23. Wu, Z., Puigserver, P., Andersson, U., Zhang, C., Adelmant, G., Mootha, V.,
Troy, A., Cinti, S., L owell, B., Scarpulla, R. C., et al. (1999) Cell 98, 115–124.
24. Bao, W., Schmid, J. E., Goetz, A. K., Ren, H. & Dix, D. J. (2005) Reprod.
Toxicol. 19, 411–419.
25. Leighton, J. K., Brown, P., Ellis, A., Harlow, P., Harrouk, W., Pine, P. S., Robison,
T., Rosario, L. & Thompson, K. (2006) Environ. Health Perspect. 114, 573–578.
26. Ishikawa, T., Tak ano, K., Yasufuku-Tak ano, J., Fujita, T., Igarashi, T., Miura,
M. & Hata, K. (2001) Nat. Biotechnol . 19, 812.
27. Eisen, M. B., Spellman, P. T., Brown, P. O. & Botstein, D. (1998) P roc. Natl.
Acad. Sci . USA 95, 14863–14868.
28. Kanehisa, M., Goto, S., Hattori, M., Aoki-Kinoshit a, K. F., Itoh, M., Ka-
washima, S., Katayama, T., Araki, M. & Hirakawa, M. (2006) Nucleic Acids Res.
34, D354–D357.
12038
www.pnas.orgcgidoi10.1073pnas.0605341103 Shioda et al.
... Several microarray studies have analyzed the transcriptomic response of cancers, specifically breast cancer, to different modalities of targeting glucose metabolism. Publications experimenting with anti-glycolytic approaches (including 2-DG, MET, and GS) have shown upregulation [23][24][25][26] or downregulation [27][28][29][30][31][32][33][34][35][36] in recurrent pathways and functions, including cell cycle; DNA replication, damage, repair; chromosome organization; nuclear division; and antigen processing and presentation via Major histocompatibility complex class I (MHC class I). ...
Article
Full-text available
Background Breast cancer (BC) is the most frequently diagnosed cancer in women. Altering glucose metabolism and its effects on cancer progression and treatment resistance is an emerging interest in BC research. For instance, combining chemotherapy with glucose-lowering drugs (2-deoxyglucose (2-DG), metformin (MET)) or glucose starvation (GS) has shown better outcomes than with chemotherapy alone. However, the genes and molecular mechanisms that govern the action of these glucose deprivation conditions have not been fully elucidated. Here, we investigated the differentially expressed genes in MCF-7 and MDA-MB-231 BC cell lines upon treatment with glucose-lowering drugs (2-DG, MET) and GS using microarray analysis to study the difference in biological functions between the glucose challenges and their effect on the vulnerability of BC cells. Methods MDA-MB-231 and MCF-7 cells were treated with 20 mM MET or 4 mM 2-DG for 48 h. GS was performed by gradually decreasing the glucose concentration in the culture medium to 0 g/L, in which the cells remained with fetal bovine serum for one week. Expression profiling was carried out using Affymetrix Human Clariom S microarrays. Differentially expressed genes were obtained from the Transcriptome Analysis Console and enriched using DAVID and R packages. Results Our results showed that MDA-MB-231 cells were more responsive to glucose deprivation than MCF-7 cells. Endoplasmic reticulum stress response and cell cycle inhibition were detected after all three glucose deprivations in MDA-MB-231 cells and only under the metformin and GS conditions in MCF-7 cells. Induction of apoptosis and inhibition of DNA replication were observed with all three treatments in MDA-MB-231 cells and metformin-treated MCF-7 cells. Upregulation of cellular response to reactive oxygen species and inhibition of DNA repair mechanisms resulted after metformin and GS administration in MDA-MB-231 cell lines and metformin-treated MCF-7 cells. Autophagy was induced after 2-DG treatment in MDA-MB-231 cells and after metformin in MCF-7 cells. Finally, inhibition of DNA methylation were observed only with GS in MDA-MB-231 cells. Conclusion The procedure used to process cancer cells and analyze their expression data distinguishes our study from others. GS had the greatest effect on breast cancer cells compared to 2-DG and MET. Combining MET and GS could restrain both cell lines, making them more vulnerable to conventional chemotherapy.
... These results show that various BPA effects are different from those of EE, while some are similar (Fig. 7). This is not surprising, since not all estrogenic substances produce the same effects [73,90,91]. Consistent with these data showing non-monotonic and more pronounced effects at low doses, the CLARITY-BPA Core study found a significant increase in mammary gland adenocarcinomas and adenomas with the lowest dose of BPA (2.5 μg/kg/day) when exposure stopped at PND 21. ...
Article
“Consortium Linking Academic and Regulatory Insights on BPA Toxicity” (CLARITY-BPA) was a comprehensive “industry-standard” Good Laboratory Practice (GLP)-compliant 2-year chronic exposure study of bisphenol A (BPA) toxicity that was supplemented by hypothesis-driven independent investigator-initiated studies. The investigator-initiated studies were focused on integrating disease-associated, molecular, and physiological endpoints previously found by academic scientists into an industry standard guideline-compliant toxicity study. Thus, the goal of this collaboration was to provide a more comprehensive dataset upon which to base safety standards and to determine whether industry-standard tests are as sensitive and predictive as molecular and disease-associated endpoints. The goal of this report is to integrate the findings from the investigator-initiated studies into a comprehensive overview of the observed impacts of BPA across the multiple organs and systems analyzed. For each organ system, we provide the rationale for the study, an overview of methodology, and summarize major findings. We then compare the results of the CLARITY-BPA studies across organ systems with the results of previous peer-reviewed studies from independent labs. Finally, we discuss potential influences that contributed to differences between studies. Developmental exposure to BPA can lead to adverse effects in multiple organs systems, including the brain, prostate gland, urinary tract, ovary, mammary gland, and heart. As published previously, many effects were at the lowest dose tested, 2.5μg/kg /day, and many of the responses were non-monotonic. Because the low dose of BPA affected endpoints in the same animals across organs evaluated in different labs, we conclude that these are biologically – and toxicologically – relevant.
... EE2 at doses equivalent to those in mixed oral contraceptives (0.4 mg/kg/day), and a 10-fold lower dose (0.04 mg/kg/day), caused many, but not all, of these same effects. While this could suggest that some effects of BPA may not be due only to its known estrogenic activity [8], it is well recognized that effects of different estrogenic chemicals on gene expression are dose dependent, and with only two doses per chemical, it is not possible to conclude that BPA and EE2 are causing effects through different response mechanisms [27]. Additionally, both BPA and EE2 elevated DNA methylation levels (5-mC) on Esr1 exon 1A and 1C (Fig. 4A). ...
Article
Full-text available
Fetal/neonatal environmental estrogen exposures alter developmental programing of the prostate gland causing onset of diseases later in life. We have previously shown in vitro that exposures to 17β-estradiol (E2) and the endocrine disrupting chemical bisphenol A, at concentrations relevant to human exposure, cause an elevation of estrogen receptor α (Esr1) mRNA in primary cultures of fetal mouse prostate mesenchymal cells; a similar result was observed in the fetal rat urogenital sinus. Effects of these chemicals on prostate mesenchyme in vivo are not well understood. Here we show effects in mice of fetal exposure to the estrogenic drug in mixed oral contraceptives, 17α-ethinylestradiol (EE2), at a concentration of EE2 encountered by human embryos/fetuses whose mothers become pregnant while on EE2-containing oral contraceptives, or bisphenol A at a concentration relevant to exposures observed in human fetuses in vivo. Expression of Esr1 was elevated by bisphenol A or EE2 exposures, which decreased the global expression of DNA methyltransferase 3A (Dnmt3a), while methylation of Esr1 promoter was significantly increased. These results show that exposures to the environmental estrogen bisphenol A and drug EE2 cause transcriptional and epigenetic alterations to expression of estrogen receptors in developing prostate mesenchyme in vivo.
... However, an epidemiological study performed in Japan found a significant association of moderate (27-51 g/day) and high (>51 g/day) soy intake with a decreased risk of NHLs in women (39). A similar inverse association between soy intake and NHL incidence was found in men in a Swedish study (40) The lack of effects at these low doses of BPA in comparison to the 50 μg/kg BW/day exposure argues against a nonmonotonic dose response curve, which has been proposed to exist for BPA according to several in vitro studies (22, [42][43][44]. With regard to the nonmonotonic effects of BPA on tumor cells in vivo, low, but not high doses of BPA were shown to significantly affect mammary tumorigenesis in a mice model (45). ...
Article
Full-text available
Well-defined physiological functions of estrogens are mediated via nuclear estrogen receptors α (ESR1) and β (ESR2). With regard to hematological malignancies, expression of ESR2 has been found in both B and T cell lymphomas. In addition to endogenous estrogens or selective ESR2 agonists, ESR2 signaling may be affected by both environmental synthetic estrogen-mimicking compounds and dietary phytoestrogens. In the present study, we demonstrate that oral exposure with either the synthetic compound bisphenol A (BPA) or the dietary phytoestrogen genistein reduced the growth of grafted murine T cell (EG7) and human B cell (Granta-519 mantle cell) lymphomas which both express ESR2. Suppression of lymphoma growth was due to reduced proliferation (BPA and genistein) and induction of apoptosis (genistein). Inhibition of lymphoma growth was seen at a BPA dose of 50 μg/kg body weight (BW)/day considered to be safe human exposure dose or a genistein dose of 1 mg/kg BW/day orally, which is reached in soy-rich diets. Thus, our study indicates that the environmental xenoestrogens BPA and genistein have anti-proliferative effects on ESR2-expressing lymphomas. Our data suggest that phytoestrogens may be considered as a dietary supplement for lymphoma patients and possibly for prevention of lymphoid malignancies.
... This notion is consistent with the reported importance of the normal development of BAT during a critical developmental window of mice to prevent obesity (49). In this context, it is interesting that our previous study detected remarkable upregulation of the mRNA transcripts encoding mitochondrial uncoupling proteins UCP1, UCP2, and UCP3 in MCF-7 human breast cancer cell culture exposed to various estrogenic endocrine disruptors, including BPA or phytoestrogens (genistein and daidzein), but not to 17b-estradiol at marker gene-standardized doses (50), suggesting possible effects of these endocrine disruptors on the development of BAT. To obtain further insights into the possible physiological dichotomy of the white and beige state of WAT and how BPA affects such developmental programming, it will be important to elucidate the molecular mechanisms of the stochastic dichotomy of the fggy TSS in future studies. ...
Article
Exposure of mammalian fetuses to endocrine disruptors can increase the risk of adult-onset diseases. For example, we previously showed that exposure of mouse fetuses to bisphenol A (BPA) caused adult-onset obesity. To obtain insights into roles of epigenetic changes in the delayed toxic effects of endocrine disruption, we determined effects of fetal mouse exposure to BPA on genome-wide DNA methylation and mRNA expression in gonadal white adipose tissues by deep sequencing (MBD-seq and RNA-seq), bisulfite pyrosequencing, and real-time qPCR. Pregnant CD-1 mice (F0) were dosed daily with 0, 5, or 500 μg/kg/day BPA during 9-18 dpc, and the weaned F1 animals were fed ad libitum with standard chow until they were sacrificed at 19 weeks old. In the vehicle-exposed F1 animals, fggy promoter showed a clear bimodal pattern of very strong (55-95%) or very weak (5-30%) DNA methylation occurring at nearly equal incidence with no intermediate strength, and promoter hypermethylation completely suppressed mRNA expression. BPA exposure eliminated this naturally occurring dichotomy, shifting fggy promoter towards the hypomethylation state to release transcriptional suppression. Strength of Fggy mRNA expression significantly correlated with increased whole-body weight and gonadal fat weight of males but not females. Bioinformatics studies showed that expression of Fggy mRNA is stronger in mouse white adipose tissues than brown adipose tissues and enhanced in gonadal fat by diet-induced obesity. These observations suggest that prenatal exposure to BPA may disrupt the physiological bimodal nature of epigenetic regulation of fggy in mouse white adipose tissues, possibly contributing to the adult-onset obesity phenotype.
... a proof of principle experiment was performed with the positive control compounds-FCCP, CCCP, and TCP and BaP as a non-uncoupling negative control. To ensure the comparability of the gene expression profiles, the test compounds were applied at equipotent concentrations (Shioda et al., 2006). The chemicals FCCP, CCCP, and TCP were tested at their EC 50 values in the TMRM assay, i.e., the concentration which inhibited TMRM uptake by 50%. ...
Article
Full-text available
A criterion frequently used to group chemicals in risk assessment is “mode of toxic action” (MoA). Routinely, structure-based approaches are used for the MoA categorization of chemicals, but they can produce conflicting results or fail to classify compounds. Biological activity-based approaches such as toxicogenomics which provide an unbiased overview of the transcriptomic changes after exposure to a compound may complement structure-based approaches in MoA assignment. Here, we investigate whether toxicogenomic profiles as generated after in vitro exposure of an established cell line (C3A hepatoma cells) are able to group together chemicals with an uncoupling MoA, and to distinguish the uncouplers from chemicals with other MoAs. In a first step, we examined whether chemicals sharing the same uncoupling of oxidative phosphorylation (OXPHOS) MoA produce similar toxicogenomic profiles and can be grouped together. In a next step, we tested whether the toxicogenomic profiles discriminate between OXPHOS and chemicals displaying a (polar) narcotic MoA. Experimentally, cells were exposed in vitro to equipotent concentrations of the test compounds and gene expression profiles were measured. The resulting toxicogenomic profiles assigned OXPHOS to one cluster and discriminated between the OXPHOS and the (polar) narcotics. In addition, the toxicogenomics data revealed that one and the same chemical can display multiple MoAs, which may help to explain conflicting results of MoA classification from structure-based approaches. The results strongly suggest the feasibility of MoA grouping of chemicals by using in vitro cell assay-based toxicogenomic profiles.
... GEO is a publicly available data repository where researchers deposit and retrieve microarray and other functional genomics data. We combined 13 transcriptome-wide datasets (Affymetrix Human Genome U133 Plus 2.0 Array only) from NCBI's GEO database describing MCF-7 cells treated with endocrine disrupting chemicals (GSE5200: [44]; GSE7765: [22]; GSE50705: [45]). Chemicals include Bisphenol A (BPA), daidzein, diethylstilbestrol (DES), 17βestradiol (E2), ethinyl estradiol (EE2), genistein, p-nonyl phenol (PNP), tris 4hydroxyphenyl-4-propyl-1-pyrazole (PPT), and dioxin. ...
Article
Targeted gene lists have been used in clinical settings to specify breast tumor type, and to predict breast cancer prognosis and response to treatment. Separately, panels have been curated to predict systemic toxicity and xenoestrogen activity as a part of chemical screening strategies. However, currently available panels do not specifically target biological processes relevant to breast development and carcinogenesis. We have developed a gene panel called the Breast Carcinogen Screen (BCScreen) as a tool to identify potential breast carcinogens and characterize mechanisms of toxicity. First, we used four seminal reviews to identify 14 key characteristics of breast carcinogenesis, such as apoptosis, immunomodulation, and genotoxicity. Then, using a hybrid data and knowledge-driven framework, we systematically combined information from whole transcriptome data from genomic databases, biomedical literature, the CTD chemical-gene interaction database, and primary literature review to generate a panel of 500 genes relevant to breast carcinogenesis. We used normalized pointwise mutual information (NPMI) to rank genes that frequently co-occurred with key characteristics in biomedical literature. We found that many genes identified for BCScreen were not included in prognostic breast cancer or systemic toxicity panels. For example, more than half of BCScreen genes were not included in the Tox21 S1500+ general toxicity gene list. Of the 230 that did overlap between the two panels, representation varied across characteristics of carcinogenesis ranging from 21% for genes associated with epigenetics to 82% for genes associated with xenobiotic metabolism. Enrichment analysis of BCScreen identified pathways and processes including response to steroid hormones, cancer, cell cycle, apoptosis, DNA damage and breast cancer. The biologically-based systematic approach to gene prioritization demonstrated here provides a flexible framework for creating disease-focused gene panels to support discovery related to etiology. With validation, BCScreen may also be useful for toxicological screening relevant to breast carcinogenesis.
Article
Background Existing cross-sectional studies indicated a positive association of bisphenol A (BPA) with overweight and obesity. However, the relationship and potential mechanisms underlying this association remain to be elucidated in prospective studies. Objective This study was designed to investigate whether serum BPA is associated with incident overweight and obesity risk, and to further explore whether adiponectin plays a mediating role in the association. Methods We measured blood BPA and adiponectin in Chinese populations. The association of serum BPA with overweight and obesity risk was evaluated using multivariable logistic regression models. We further examined the mediating effect of adiponectin by causal mediation analysis. Results Among 796 participants free of overweight and obesity at baseline, 133 individuals developed overweight and obesity during the follow-up period. Compared with those in the lowest quartile of serum BPA, those in the second and third quartiles were positively associated with incident overweight and obesity risk adjusting for covariates (all P-values < 0.05), whereas this association was not observed in the fourth quartile. Further spline analysis showed an inverted U-shaped dose-response relationship (Pnon-linear = 0.04). Furthermore, each unit of serum log10-transformed BPA levels was associated with higher changes in waist-to-height ratio and body roundness index (all P-values < 0.05). Mediation analysis indicated significant indirect effects of adiponectin on the associations of BPA with overweight and obesity prevalence (mediation proportion: 46.08%; P = 0.02), and BMI levels (mediation proportion: 30.32%; P = 0.03). Conclusion Serum BPA displayed a positive association with incident overweight and obesity risk in a non-monotonic pattern, and adiponectin might mediate the association. Further mechanistic studies are warranted.
Chapter
There are a number of features of endocrine-disrupting chemicals (EDCs) that distinguish them from traditional toxicants. One such attribute is the ability of EDCs to induce nonmonotonic dose response curves, where low and high doses can produce opposite effects. Another is that EDCs can disrupt hormone-sensitive outcomes, even when exposures are very low, e.g., administered in levels as low as nanogram or microgram per kilogram in experimental animals, or the low doses that humans encounter in their everyday lives. This chapter describes the mechanisms by which low dose effects and nonmonotonic responses can manifest and some specific examples from the EDC literature. It also describes the recent debate over whether these effects are “real,” and whether they are common enough to influence chemical safety assessments for EDCs. Together, these features of EDCs suggest that the methods used to determine safe doses for human exposures are flawed.
Article
The development and function of the mammary gland are endocrine-dependent processes, depending on the stage of development. Foetal and/or postnatal exposure to low doses of BPA alters tissue organisation through epithelial proliferation and stroma-epithelial interactions. BPA also alters the expression of E2-dependent epithelial and stroma transcriptomes. Several signalling pathways are consistent with the observed phenotype: proliferation and apoptosis, a focal adhesion pathway indicating changes in biomechanical properties of the extracellular matrix, and immune function. Some of BPA's effects are reversed by oestrogen and/or GPER inhibitors. BPA also alters the expression of epigenetic marks (EZH2, HOTAIR), which would explain the delayed effect of foetal BPA exposure. In conclusion, experimental evidence shows that pre- or postnatal BPA exposure consistently causes endocrine modifications in the mammary tissue of different animal species, disrupting stromal-epithelial interactions and ultimately increasing its susceptibility to carcinogens. An interspecies comparison highlights why and how these effects apply to humans.
Article
Full-text available
Alkylphenols perturb the endocrine system and are considered to have weak estrogenic activities. Although it is known that nonylphenol can bind weakly to the estrogen receptor, it is unclear whether all reported effects of nonylphenol are attributable to its estrogen receptor-binding activity. In order to examine whether alkylphenols have similar effects to the natural hormone, estradiol, we used a mouse model to examine the effects of nonylphenol on gene expression and compared it with estradiol. DNA microarray analysis revealed that, in the uterus, most of the genes activated by this alkylphenol at a high dose (50 mg/kg) were also activated by estradiol. At lower doses, nonylphenol (0.5 mg/kg and 5 mg/kg) had little effect on the genes that were activated by estradiol. Thus, we concluded that the effects of nonylphenol at a high dose (50 mg/kg) were very similar to estradiol in uterine tissue. Moreover, since evaluation of estrogenic activity by gene expression levels was comparable with the uterotrophic assay, it indicated that analysis of gene expression profiles can predict the estrogenic activities of chemicals. In contrast to the similar effects of nonylphenol and estradiol observed in the uterus, in the liver, gene expression was more markedly affected by nonylphenol than by estradiol. This indicated that, in the liver, nonylphenol could activate another set of genes that are distinct from estrogen-responsive genes. These results indicated that nonylphenol has very similar effects to estradiol on gene expression in uterine but not in liver tissue, indicating that tissue-specific effects should be considered in order to elucidate the distinct effects of alkylphenols.
Article
Full-text available
The steroid hormone estrogen can stimulate mitogenesis in hormone-responsive breast cancer epithelial cells. This action is attributed to the transcriptional activity of the ER, a ligand-dependent transcription factor. However, the exact molecular mechanism underlying estrogen-induced proliferation has yet to be completely elucidated. Using custom cDNA microarrays containing many genes implicated in cell cycle progression and DNA replication, we examined the gene expression of a hormone-responsive breast cancer cell line (MCF-7) treated with a mitogenic dose of estrogen in the absence of confounding growth factors found in serum. Gene expression changes were monitored 1, 4, 12, 24, 36, and 48 h after estrogen stimulation so that RNA levels at critical times throughout cell cycle progression could be monitored. Significant changes include the altered transcript levels of genes implicated in transcription, cellular signaling, and cell cycle checkpoints. At time points during which increased numbers of cells were progressing through S phase, a majority of the genes associated with the DNA replication fork were also found to be induced. The coexpression of DNA replication fork genes by estrogen without the support of serum growth factors indicates an important estrogen regulatory component of the molecular mechanism driving estrogen-induced mitogenesis.
Article
Full-text available
Genomic sequencing has made it clear that a large fraction of the genes specifying the core biological functions are shared by all eukaryotes. Knowledge of the biological role of such shared proteins in one organism can often be transferred to other organisms. The goal of the Gene Ontology Consortium is to produce a dynamic, controlled vocabulary that can be applied to all eukaryotes even as knowledge of gene and protein roles in cells is accumulating and changing. To this end, three independent ontologies accessible on the World-Wide Web (http://www.geneontology.org) are being constructed: biological process, molecular function and cellular component.
Article
Alkylphenols perturb the endocrine system and are considered to have weak estrogenic activities. Although it is known that nonylphenol can bind weakly to the estrogen receptor, it is unclear whether all reported effects of nonylphenol are attributable to its estrogen receptor-binding activity. In order to examine whether alkylphenols have similar effects to the natural hormone, estradiol, we used a mouse model to examine the effects of nonylphenol on gene expression and compared it with estradiol. DNA microarray analysis revealed that, in the uterus, most of the genes activated by this alkylphenol at a high dose (50 mg/kg) were also activated by estradiol. At lower doses, nonylphenol (0·5 mg/kg and 5 mg/kg) had little effect on the genes that were activated by estradiol. Thus, we concluded that the effects of nonylphenol at a high dose (50 mg/kg) were very similar to estradiol in uterine tissue. Moreover, since evaluation of estrogenic activity by gene expression levels was comparable with the uterotrophic assay, it indicated that analysis of gene expression profiles can predict the estrogenic activities of chemicals. In contrast to the similar effects of nonylphenol and estradiol observed in the uterus, in the liver, gene expression was more markedly affected by nonylphenol than by estradiol. This indicated that, in the liver, nonylphenol could activate another set of genes that are distinct from estrogen-responsive genes. These results indicated that nonylphenol has very similar effects to estradiol on gene expression in uterine but not in liver tissue, indicating that tissue-specific effects should be considered in order to elucidate the distinct effects of alkylphenols.
Article
In order to understand early events caused by estrogen in vivo, temporal uterine gene expression profiles at early stages were examined using DNA microarray analysis. Ovariectomized mice were exposed to 17β-estradiol and the temporal mRNA expression changes of ten thousand various genes were analyzed. Clustering analysis revealed that there are at least two phases of gene activation during the period up to six hours. One involved immediate-early genes, which included certain transcription factors and growth factors as well as oncogenes. The other involved early-late genes, which included genes related to RNA and protein synthesis. In clusters of down-regulated genes, transcription factors, proteases, apoptosis and cell cycle genes were found. These hormone-inducible genes were not induced in estrogen receptor (ER) α knockout mice. Although expression of ERβ is known in the uterus, these findings indicate the importance of ERα in the changes in gene expression in the uterus.
Article
In order to understand early events caused by estrogen in vivo, temporal uterine gene expression profiles at early stages were examined using DNA microarray analysis. Ovariectomized mice were exposed to 17β-estradiol and the temporal mRNA expression changes of ten thousand various genes were analyzed. Clustering analysis revealed that there are at least two phases of gene activation during the period up to six hours. One involved immediate-early genes, which included certain transcription factors and growth factors as well as oncogenes. The other involved early-late genes, which included genes related to RNA and protein synthesis. In clusters of down-regulated genes, transcription factors, proteases, apoptosis and cell cycle genes were found. These hormone-inducible genes were not induced in estrogen receptor (ER) α knockout mice. Although expression of ERβ is known in the uterus, these findings indicate the importance of ERα in the changes in gene expression in the uterus.
Article
Estrogen plays an important role in many physiological events including carcinogenesis and the development of human breast cancer. However, the molecular mechanisms of estrogen signaling in cancers have not been clarified hitherto and accurate therapeutic prediction of breast cancer is earnestly desired. We first carried out estrogen-responsive expression profiling of approximately 9000 genes in estrogen receptor-positive human MCF-7 breast cancer cells. Based on the results, estrogen-responsive genes were selected for production of a custom-made cDNA microarray. Using a microarray consisting of the narrowed-down gene subset, we first analyzed the time course of the estrogen-responsive gene expression profiles in MCF-7 cells, resulting in subdivision of the genes up-regulated by estrogen into early-responsive and late-responsive genes. The expression patterns of several genes were confirmed by Northern blot analysis. We also analyzed the effects of the estrogen antagonists ICI 182780 and 4-hydroxytamoxifen (OHT) on the estrogen-responsive gene expression profiles in MCF-7 cells. While the regulation of most of the genes by estrogen was completely abolished by ICI 182780, some genes were partially regulated by estrogen even in the presence of OHT. Furthermore, the estrogen-responsive gene expression profiles of twelve cancer cell lines derived from the breast, ovary, stomach and other tissues were obtained and analyzed by hierarchical clustering including the profiles in MCF-7 cells. Several genes also showed up-regulation or down-regulation by estrogen in cell lines other than MCF-7 cells. The significance of the estrogen-responsive genes identified in these analyses concerning the nature of cancer is discussed.
Article
A system of cluster analysis for genome-wide expression data from DNA microarray hybridization is described that uses standard statistical algorithms to arrange genes according to similarity in pattern of gene expression. The output is displayed graphically, conveying the clustering and the underlying expression data simultaneously in a form intuitive for biologists. We have found in the budding yeast Saccharomyces cerevisiae that clustering gene expression data groups together efficiently genes of known similar function, and we find a similar tendency in human data. Thus patterns seen in genome-wide expression experiments can be interpreted as indications of the status of cellular processes. Also, coexpression of genes of known function with poorly characterized or novel genes may provide a simple means of gaining leads to the functions of many genes for which information is not available currently.
Article
Mitochondrial number and function are altered in response to external stimuli in eukaryotes. While several transcription/replication factors directly regulate mitochondrial genes, the coordination of these factors into a program responsive to the environment is not understood. We show here that PGC-1, a cold-inducible coactivator of nuclear receptors, stimulates mitochondrial biogenesis and respiration in muscle cells through an induction of uncoupling protein 2 (UCP-2) and through regulation of the nuclear respiratory factors (NRFs). PGC-1 stimulates a powerful induction of NRF-1 and NRF-2 gene expression; in addition, PGC-1 binds to and coactivates the transcriptional function of NRF-1 on the promoter for mitochondrial transcription factor A (mtTFA), a direct regulator of mitochondrial DNA replication/transcription. These data elucidate a pathway that directly links external physiological stimuli to the regulation of mitochondrial biogenesis and function.