ArticlePDF Available

Effects of Exposure to the Endocrine-Disrupting Chemical Bisphenol A During Critical Windows of Murine Pituitary Development

Authors:

Abstract and Figures

Critical windows of development are often more sensitive to endocrine disruption. The murine pituitary gland has two critical windows of development: embryonic gland establishment and neonatal hormone cell expansion. During embryonic development, one environmentally ubiquitous endocrine disrupting chemical (EDC), bisphenol A (BPA), has been shown to alter pituitary development by increasing proliferation and gonadotrope number in females, but not males. However, the effects of exposure during the neonatal period were not examined. Therefore, in this study, we dosed pups from postnatal day (PND) 0 to PND7 with 0.05, 0.5, and 50μg/kg/day BPA, environmentally relevant doses, or 50μg/kg/day estradiol (E2). Mice were collected after dosing at PND7 and at five weeks. Neonatally dosing mice with BPA caused sex specific gene expression changes distinct from those observed with embryonic exposure. At PND7, pituitary Pit1 mRNA expression was decreased with BPA 0.05 and 0.5μg/kg/day in males only. Expression of Pomc mRNA was decreased at 0.5μg/kg/day BPA in males and 0.5, 50μg/kg/day BPA in females. Similarly, E2 decreased Pomc mRNA in both males and females. However, there were no noticeable corresponding changes in protein expression. Both E2 and BPA suppressed Pomc mRNA in pituitary organ cultures and this repression appeared to be mediated by ESR1 and ESR2 in females and by GPER in males, as determined by estrogen receptor subtype-selective agonists. These data demonstrate that BPA exposure during neonatal pituitary development has unique sex specific effects on gene expression and Pomc repression in males and females may occur through different mechanisms.
Content may be subject to copyright.
RESEARCH ARTICLE
Effects of Exposure to the Endocrine-Disrupting
Chemical Bisphenol A During Critical Windows
of Murine Pituitary Development
Kirsten S. Eckstrum,
1
Whitney Edwards,
1
Annesha Banerjee,
1
Wei Wang,
2
Jodi A. Flaws,
2
John A. Katzenellenbogen,
3
Sung Hoon Kim,
3
and Lori T. Raetzman
1
1
Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana,
Illinois 61801;
2
Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801;
and
3
Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
Critical windows of development are often more sensitive to endocrine disruption. The murine
pituitary gland has two critical windows of development: embryonic gland establishment and
neonatal hormone cell expansion. During embryonic development, one environmentally ubiquitous
endocrine-disrupting chemical, bisphenol A (BPA), has been shown to alter pituitary development
by increasing proliferation and gonadotrope number in females but not males. However, the effects
of exposure during the neonatal period have not been examined. Therefore, we dosed pups from
postnatal day (PND)0 to PND7 with 0.05, 0.5, and 50 mg/kg/d BPA, environmentally relevant doses, or
50 mg/kg/d estradiol (E2). Mice were collected after dosing at PND7 and at 5 weeks. Dosing mice
neonatally with BPA caused sex-specific gene expression changes distinct from those observed with
embryonic exposure. At PND7, pituitary Pit1 messenger RNA (mRNA) expression was decreased with
BPA 0.05 and 0.5 mg/kg/d in males only. Expression of Pomc mRNA was decreased at 0.5 mg/kg/d BPA
in males and at 0.5 and 50 mg/kg/d BPA in females. Similarly, E2 decreased Pomc mRNA in both males
and females. However, no noticeable corresponding changes were found in protein expression.
Both E2 and BPA suppressed Pomc mRNA in pituitary organ cultures; this repression appeared to be
mediated by estrogen receptor-aand estrogen receptor-bin females and G proteincoupled es-
trogen receptor in males, as determined by estrogen receptor subtype-selective agonists. These data
demonstrated that BPA exposure during neonatal pituitary development has unique sex-specific
effects on gene expression and that Pomc repression in males and females can occur through
different mechanisms. (Endocrinology 159: 119131, 2018)
The pituitary is the master gland of the endocrine
system, releasing hormones that control reproduc-
tion, lactation, growth, metabolism, and stress. To re-
lease these hormones, the anterior pituitary gland must
develop five hormone-secreting cell types: gonadotropes,
lactotropes, somatotropes, thyrotropes, and cortico-
tropes. Development of the hormone-secreting cell types
occurs during two key periods in mouse development:
embryonic and neonatal. During embryonic develop-
ment, cell specification is regulated by intrinsic and
paracrine signals from the pituitary and surrounding
tissue (1). However, during the neonatal period, the
capability for communication between the hypothala-
mus, pituitary gland, and target organs develops (2, 3),
suggesting a potential role for axis hormones in
this window of development. Additionally, at birth,
the testosterone surge establishes sex differences in
the hypothalamicpituitarygonadal (HPG) and the
hypothalamicpituitaryadrenal (HPA) axes through
local aromatization of testosterone to estradiol in the
ISSN Online 1945-7170
Copyright © 2018 Endocrine Society
Received 16 June 2017. Accepted 3 October 2017.
First Published Online 6 October 2017
Abbreviations: ACTH, adrenocorticotropic hormone; BPA, bisphenol A; BPA0.05,
0.05 mg/kg/d bisphenol A; BPA0 .5, 0 .5 mg/kg/d bisphenol A; BPA50, 50 mg/kg/d
bisphenol A; CAS, Chemical Abstracts Servi ce; DPN, diarylpropionitrile; E2, estradiol;
EDC, endocrine-disrupting chemical; ESR1, estrogen receptor-a; ESR2, estrogen
receptor-b; GnRH, gonadotropin-releasing hormone; GPER, G proteincoupled
estrogen receptor; HPA, hypothalamicpituitaryadrenal; HPG, hypothalamic
pituitarygonadal; IHC, immunohistochemical; PBS, phosphate-buffered saline;
PND, postnatal day; PPT, pyrazole triol.
doi: 10.1210/en.2017-00565 Endocrinology, January 2018, 159(1):119131 https://academic.oup.com/endo 119
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
male brain (4, 5). Thus, the endocrine milieu postnatally
can alter pituitary sensitivity to exposure to endocrine-
disrupting chemicals (EDCs).
Bisphenol A (BPA) is an EDC found in several items
such as polycarbonate plastics, epoxy resins, and thermal
paper, leading to ubiquitous exposure (6). Studies have
suggested that the developing organs of fetuses and ne-
onates have heightened sensitivity to EDCs such as di-
ethylstilbestrol and BPA in both rodents and humans
(79). BPA is readily detectible in fetal cord serum,
maternal serum, and fetal amniotic fluid in humans (10),
demonstrating exposure to BPA during critical devel-
opmental windows. A possible mechanism by which
BPA is considered to act is by disrupting the actions of
estrogens. The classic estrogen receptors estrogen receptor-
a(ESR1) and estrogen receptor-b(ESR2) and the mem-
brane G proteincoupledestrogenreceptor(GPER)are
found in the hypothalamus and pituitary gland (1115).
However, the roles of estrogen signaling and the potential
effects of BPA during neonatal pituitary development are
unexplored.
Developmental exposure to environmentally relevant
doses of BPA at or less than the no observed adverse effect
level can affect every axis of the endocrine system reg-
ulated by the pituitary gland, including the reproductive
axis (1620) in rats and mice and the prolactin levels (21),
growth axis (22), thyroid axis (23), and stress axis (24) in
rats. BPA also alters sex differences of the HPG and HPA
axes established by sex steroids (2427). Despite the
relatively quick metabolism of BPA (28), changes can
sometimes occur well after exposure has ended. For
example, embryonic exposure to 25 to 250 ng/kg/d BPA
led to reproductive tissue weight changes in adult female
mice and changes in estrogen and progesterone receptor
expression (9). Also, neonatal exposure to 50 mg/kg/d to
50 mg/kg/d BPA had different outcomes on the rat hy-
pothalamus at 2 days after exposure than at 8 days after
exposure (27), indicating the importance of examinations
at multiple points after exposure. Therefore, it is clear
that developmental exposure to BPA can influence sex
differences and the endocrine axes, although regulation
could be time and/or context specific. Despite this evi-
dence, few studies have examined closely the effects of
neonatal BPA exposure on the development and ex-
pression of genes in the pituitary gland itself.
Previously, our laboratory showed that embryonic
exposure of pregnant mice to 0.5 or 50 mg/kg/d BPA
increased pituitary proliferation and gonadotrope cell
numbers in female offspring (29), a response similar to
that of estradiol (E2) (30). However, the effects of neonatal
exposure on the second wave of pituitary development are
unknown. We hypothesized that BPA exposure would
have different effects on the pituitary gland, depending on
sex, the developmental period of exposure, and the timeof
examination after exposure. Additionally, we hypothe-
sized that some of these effects might be similar to that of
E2. To test this hypothesis, two experiments were per-
formed. First, we examined pituitary glands at two time
points after exposure neonatally to three environmentally
relevant doses of BPA and E2 and compared the effects in
males and females. Second, we treated isolated pituitary
glands in culture with E2, BPA, and estrogen receptor
subtype-specific agonists to determine the direct effects on
the pituitary gland and an estrogen receptor pathway
important in gene regulation.
Materials and Methods
Mice
CD-1 mice, obtained from Charles River, were bred in-
house, kept in polysulfone cages containing corn cob bedding
material, and fed Teklad 8664 rodent chow (Envigo) and water
ad libitum through glass bottles. Sex was confirmed by visual
inspection, SRY genotyping as previously described (31), or
gonadal removal. The University of Illinois Urbana-Champaign
institutional animal care and use committee approved all
procedures.
Experiment 1 design: effects of neonatal dosing
In the first experiment, we examined the effects of dosing
male and female mice during the critical neonatal period of
pituitary development. CD-1 litters were culled to 8 to 12 pups
on the day of birth, and the neonatal pups were dosed orally
through consumption from a pipet tip, once daily, from post-
natal day (PND)0 to PND7 with control (0.1% ethyl alcohol),
0.05, 0.5, or 50 mg/kg/d BPA [BPA0.05, BPA0.5, and BPA50,
respectively; Sigma-Aldrich; purity, $99%; Chemical Abstracts
Service (CAS) no. 80-05-7], or 50 mg/kg/d E2 (17b-estradiol;
Tocris; purity, .99%; CAS no. 50-28-2) dissolved in tocopherol-
stripped corn oil (MP Biomedicals). The litters were randomly
assigned to a treatment group. The 0.05- and 0.5-mg/kg/d doses of
BPA were chosen because they are within the range of human
exposure levels (32). The 50-mg/kg/d dose of BPA was chosen
because it is the oral reference dose for BPA (33). The 50-mg/kg/d
dose of E2 was chosen because it was a dose sufficient to induce
expression of estrogen-regulated genes in our system (31). All
pups in a litter were dosed with a single compound, and six
separate litters were dosed with each compound. One male and
one female were taken from each litter at each measurement point
for each assay between 10 and 11 AM.
Part A: on neonatal pituitary development
Part A of the experiment examined the immediate effects of
exposure to BPA and E2 during the neonatal period. The mice
were euthanized on PND7, 1 hour after the final dosing. For
western blot analysis, an additional dosing of each compound
was performed, and all pups were collected at PND7 for western
blot analysis.
Part B: at 5 weeks
Part B of the experiment examined whether neonatal dosing
would influence the pituitary outside the first postnatal rapid
120 Eckstrum et al Bisphenol A Effects on Postnatal Pituitary Gland Endocrinology, January 2018, 159(1):119131
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
growth phase, which ends at 3 weeks of age (34). The 5-week-old
siblings of the mice examined at PND7 were collected for RNA
analysis. One male and female were analyzed from each litter, if
possible. Because of the sex distribution, it was not always possible
to have the number equal six. The estrous cycles were monitored
beginning at 5 weeks by vaginal smear (35), and the mice were
euthanized when in estrus. Female mice were euthanized in estrus
because neonatal E2 treatment caused persistent estrus.
Measurements, weights, and puberty assessment
At PND7 and 5 weeks, the weights and measurements were
recorded to determine whether any gross systemic effects of BPA or
E2 exposure had occurred.The wet weight for the testes, ovaries,
uteri, and liver were all compared with the body weight. The
anogenital distance was measured using calipers and compared
with the body weight. Puberty measurements were observed,
stratified by the day of vaginal opening for females and preputial
separation for males.
Experiment 2 design: pituitary cultures
PND1 pituitary glands were cultured overnight on Millicell
CM membranes (Millipore), as previously described (31). The
next day, the media were replaced with media containing the
following compounds alone or in combination: 10
28
M pyr-
azole triol (PPT; Tocris; purity, .99%; CAS no. 263717-53-9),
10
28
M diarylpropionitrile (DPN; Tocris; purity, .99%; CAS
no. 1428-67-7), 10
28
M E2 (Tocris; purity, .99%; CAS no. 50-
28-2), 4.4 310
25
M BPA (Sigma-Aldrich; purity, $99%; CAS
no. 80-05-7), 10
26
M G1 (Cayman; purity, .98%; CAS no.
881639-98-1), or vehicle control consisting of 0.1% ethanol for
treatment with a single compound, 0.2% ethanol for cotreat-
ment, or 0.1% dimethyl sulfoxide for G1 treatment. The pi-
tuitary glands were also treated with an estrogen dendrimer
conjugate (10
29
M) or empty dendrimer conjugate (dendrimer
control; 10
29
M) (36, 37). For the cotreatment experiments
with PPT and DPN, 0.2% ethanol vehicle was added to the
dendrimer control. The pituitary glands were treated for
48 hours before collection (n = 6 to 13).
Quantitative reverse transcription polymerase
chain reaction
For RNA analysis, males and females were analyzed separately
(n = 3 to 6). RNA was processed, as previously described (38). The
expression levels of the genes of interest were normalized to Actb
messenger RNA (mRNA) levels, with the primer sequences of
forward 50to 30: GACATGGAGAAGATCTGGCA and reverse
50to 30: GGTCTCAAACATGATCTGGGT (Life Technologies).
Gene expression for Mki67,Lhb,Nr5a1,Tshb,Gh,andPrl was
analyzed using primer sequences previously reported (29) and Pit1
and Tpit previously reported (38). Finally, the Pomc sequences
used were forward 50to 30: GATAGCGGGAGAGAAAGCCG
and reverse 50to 30: GGGACCCCGTCCTGTCCTAT. The data
were analyzed using the standard comparative change in threshold
cycle(DCt) value method, as previously described (39). For in vivo
analysis, individual points were graphed, with the sex indicated
and averages shown as a line. For pituitary cultures, bar graphs
representing the data were prepared.
Immunohistochemistry
Pituitary glands were collected at PND7 from the exposed
mice and fixed in 3.7% paraformaldehyde for 1 hour, cryoprotected
in 30% sucrose/phosphate-buffered saline (PBS) solution, and
frozen in optimal cutting temperature compound (Electron
Microscopy Sciences). The pituitary glands were sectioned into
10-mm slices before being mounted onto Superfrost plus slides
(Fisher Scientific).
Before antibody treatments, frozen sections were thawed for
10 minutes, fixed in 4% paraformaldehyde diluted in PBS, and
blocked using 5% normal donkey serum diluted in an immu-
nohistochemical (IHC) block, which had 3% bovine serum
albumin and 0.5% TritonX-100 diluted in PBS. The slides were
then treated with the following primary antibodies overnight at
4°C: LHb(luteinizing hormone-b; National Hormone and
Peptide Program), phosphorylated histone H3 (Millipore), PIT1
(gift from Simon Rhodes), and POMC (Dako). After a series of
PBS washes, the slides were treated with biotin-conjugated
rabbit secondary antibody (Jackson ImmunoResearch) diluted
1:200 in an IHC block for 60 minutes at room temperature. After
another series of PBS washes, the slides were treated with
streptavidin-conjugated cy3 tertiary antibody (Jackson Immuno-
Research) diluted 1:200 using an IHC block. Control slides were
prepared that did not include the primary antibody.
All slides were counterstained with 40,6-diamidino-2-
phenylindole (1:1000; Sigma-Aldrich) and visualized under a
Leica DM2560 microscope. Photographs were taken using a
Retiga 2000R camera (Q-Imaging) and acquired using Q-
Capture Pro software (Q-Imaging). Images were processed
using Adobe Photoshop CS2.
Cell counting
POMC cell counts were performed using National Institutes
of Health ImageJ. For each dose (0.05, 0.5, and 50 mg/kg/d BPA,
and control) four to five pituitary glands were examined, with
three to four slides per pituitary. The percentage of POMC-
positive cell numbers was determined by comparing the
number of POMC immunoreactive cells per 40,6-diamidino-2-
phenylindole stained nuclei in a defined area. The sections on
each slide, and all the slides per animal (3), were averaged
together for the mean percentage of positive POMC cells for
each mouse.
Western blot
Three pituitary glands were frozen on dry ice and lysed using
radioimmunoprecipitation assay buffer. Protein fractions were
obtained by centrifugation for 5 minutes at 4°C at 20,817
relative centrifugal force. The total protein concentration
was measured via a BCA protein assay kit (Thermo Fisher)
according to the manufacturers instructions. Protein samples
(20 mg) were mixed with radioimmunoprecipitation assay
buffer and 33Laemmli loading dye and then heated for
12 minutes at 57°C. The samples were loaded on a 10% sodium
dodecyl sulfate-polyacrylamide gel electrophoresis gel and
transferred to a nitrocellulose membrane (BioRad). The mem-
brane was blocked for 2 hours in 5% nonfat dried milk in Tris-
buffered saline and incubated with either PIT1 antibody (rabbit;
1:1000; gift from Dr. Simon Rhodes), POMC antibody (rabbit;
1:250; Dako), b-actin antibody (rabbit; 1:1000; Cell Signaling),
or a-tubulin antibody (mouse; 1:5000; Sigma-Aldrich) in 1%
milk with 0.2% Tween, overnight at 4°C. Secondary goat
anti-rabbit (1:500) or goat anti-mouse (1:5000) IgG conjugated
to IRdye 800CW (LiCor) was added at room temperature for
1 hour. The membrane was then imaged with the Odyssey IR
doi: 10.1210/en.2017-00565 https://academic.oup.com/endo 121
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
imaging system (LiCor). The relative protein levels were ana-
lyzed using National Institutes of Health ImageJ.
Statistical analysis
Statistical significance among the controls, three BPA
treatment groups, and E2 was determined using one-way
analysis of variance within sex. Within sex was used because
sex differences were expected in some gene expression levels
between the control males and females (31). Only one pituitary
gland of each sex from each litter was used in the quantitative
polymerase chain reaction (PCR) analysis; thus, litter was not
considered as a covariate. If significance was found, the mean
fold change between groups was compared using the Tukey
honest significant difference test in Microsoft Excel for both
quantitative reverse transcription PCR and cell counting. For
quantitative reverse transcription PCR of pituitary explant
culture experiments, significance was determined using a two-
tailed ttest. Pvalues of #0.05 were considered statistically
significant.
Results
Experiment 1: neonatal dosing caused few changes
in gross appearance of mice
Previous studies have demonstrated that the embryonic
pituitary gland is sensitive to BPA exposure. Therefore, we
sought to examine the effects of BPA exposure specifi-
cally during neonatal development. Organ weights and
measurements were taken to determine whether any
gross effects were present. The testes, ovaries, uteri, and
liver weights are provided as a percentage of body
weight (Table 1). At PND7, the lowest dose of BPA
decreased the ovary weight compared with the control,
and E2 had increased the uterine weight compared with
thecontrol.At5weeks,thesameweightsandmea-
surements were gathered. No statistically significant
differences were found in the body weights or mea-
surements between any of the treatments at the 5-week
point (Table 2).
Experiment 1: neonatal exposure to BPA did not
affect pituitary proliferation
Exposure to BPA or E2 during the neonatal period had
no statistically significant effect on pituitary proliferation
at PND7, as measured by Mki67 mRNA levels, in males
[Fig. 1(a); F
(4, 25)
= 1.82; P= 0.16) and females [Fig. 1(b);
F
(4, 25)
= 0.74; P= 0.57]. Immunohistochemistry for the
mitosis marker phosphorylated histone H3 in the female
[Fig. 1(e)1(h)] and male [Fig. 1(i)1(l)] pituitary glands
at PND7 revealed no obvious change in the distribution
of cells stained in response to exposure to BPA. We next
examined the proliferation at 5 weeks to determine
whether an effect of neonatal BPA exposure would be
present at a later time point. The Mki67 mRNA levels
were not significantly altered statistically by BPA or E2 in
the males [Fig. 1(c); F
(4, 17)
= 1.66; P= 0.21] or females
[Fig. 1(d); F
(4, 19)
= 1.61; P= 0.21] at 5 weeks.
Experiment 1: neonatal BPA exposure did not affect
gonadotrope lineage gene expression
To examine whether BPA exposure during the neo-
natal period affected the gonadotrope lineage, the mRNA
expression levels of the gonadotrope lineage transcription
factor Nr5a1 were examined. No statistically significant
effect of exposure on Nr5a1 mRNA levels was found in
males at PND7 [Fig. 2(a); F
(4, 25)
= 0.81; P= 0.53). In
females, a statistically significant effect of exposure was
found on Nr5a1 mRNA levels [Fig. 2(b); F
(4, 25)
= 2.85;
P#0.04]. An increase in Nr5a1 with E2 treatment was
found (P#0.01). To explore whether a difference in
Nr5a1 mRNA levels would exist later, we examined the
pituitary glands at 5 weeks. In these mice, the males
showed no statistically significant main effect of treat-
ment on Nr5a1 mRNA levels [Fig. 2(c); F
(4, 17)
= 0.94; P=
0.47]. However, in females, an effect of treatment was
seen [F
(4, 18)
= 4.92; P#0.007], and the Nr5a1 mRNA
levels were decreased with the BPA0.05 and E2 exposures
[Fig. 2(d); P#0.002 and P#7310
25
].
To further explore the gonadotrope lineage during
neonatal exposure to E2 or BPA, Lhb mRNA expression
was analyzed. With neonatal dosing, a statistically sig-
nificant effect of exposure on Lhb mRNA levels was
found in males [F
(4, 25)
= 4.04; P#0.01] and females
[F
(4, 25)
= 9.70; P#7310
25
]. BPA exposure did not
statistically significantly alter Lhb mRNA levels. How-
ever, E2 significantly decreased the levels of Lhb mRNA
Table 1. PND7 Wet Weights and Measurements
Variable
Weight, g
Testes, %BW Ovaries, %BW Uteri, %BW Liver, %BW
AGD, mm/gBW
Male Female Male Female
Control 4.725 4.417 0.175 0.049 0.093 3.118 1.14 0.91
BPA0.05 4.217 3.995 0.177 0.038
a
0.094 3.012 1.16 0.82
BPA0.5 4.611 4.250 0.170 0.043 0.096 3.137 1.10 0.83
BPA50 4.606 4.483 0.179 0.044 0.104 3.201 1.20 0.82
E2 4.683 4.555 0.183 0.057 0.195
a
3.163 1.15 0.82
Abbreviations: AGD, anogenital distance; gBW, grams of body weight; %BW, percentage of body weight.
122 Eckstrum et al Bisphenol A Effects on Postnatal Pituitary Gland Endocrinology, January 2018, 159(1):119131
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
in both males [Fig. 2(e); P#0.04] and females [Fig. 2(f);
P#0.0005] at PND7. Immunohistochemistry for LHb
did not revealany obvious differences inthe BPA treatment
groups in females (Supplemental Fig. 1). Additionally,
mRNA levels for the common a-subunit Cga were ex-
amined at PND7 and were not statistically significantly
affected by exposure in males [F
(4, 24)
=0.78;P= 0.55] or
females [F
(4, 24)
= 0.40; P= 0.81]. We next examined the
5-week-old mice. For the Lhb mRNA levels, no statis-
tically significant effect of BPA or E2 exposure was found
in males [Fig. 2(g); F
(4, 17)
= 0.78; P= 0.55]. A statistically
significant effect of exposure was found for females
[Fig. 2(h); F
(4, 19)
= 11.61; P#6310
25
]. No statistically
significant difference was found in Lhb mRNA with BPA
exposure; however, suppression of Lhb mRNA occurred
with E2 exposure [Fig. 2(h); P#6310
25
]. Despite
finding effects on the gonadotrope lineage with E2
treatment, no statistically significant effects were found
on the timing of puberty, as assessed by preputial sep-
aration or vaginal opening with E2 or BPA treatment
(Table 3).
Experiment 1: low levels of BPA exposure decreased
Pit1 mRNA in males only, without altering hormone
transcript levels
PIT1 is a transcription factor found exclusively in the
pituitary gland and is important in the development of
lactotropes, somatotropes, and thyrotropes and expres-
sion of their respective hormones. A statistically signifi-
cant effect of exposure on Pit1 mRNA levels was found in
males [Fig. 3(a); F
(4, 25)
= 3.23; P#0.03]. The lowest
doses of BPA, BPA0.05 and BPA0.5, decreased the levels
of Pit1 mRNA (P#0.002 and P#0.007, respectively);
however, BPA50 and E2 had no statistically significant
effect on Pit1 mRNA during this period. In females,
exposure did not have a statistically significant effect on
Pit1 mRNA [Fig. 3(b); F
(4, 25)
= 1.84; P= 0.15]. To
determine whether the decrease in Pit1 mRNA in males
would correspond with a decrease in protein, western
blot analysis was performed. No statistically significant
change in PIT1 protein was seen with any treatment
group in the males at PND7 [Fig. 3(c)]. Additionally,
immunohistochemistry for PIT1 did not reveal any ob-
vious differences in the treatment groups in the males
(Supplemental Fig. 1). PIT1 is a transcriptional regulator
of Gh,Prl,andTshb. Consequently, we examined
mRNA expression of these hormones to determine
whether BPA-mediated repression of Pit1 mRNA resul-
ted in downstream effects on hormone mRNA expres-
sion. A statistically significant effect of exposure was
found for Prl mRNA levels in males [F
(4, 25)
= 53.86; P#
6310
212
] and females [F
(4, 25)
= 17.81; P#1310
26
].
BPA had no substantial effect on Prl mRNA; however, the
increase in Prl mRNA with E2 was statistically significant
in males [Fig. 3(d); P#1310
25
] and females [Fig. 3(e);
P#0.0002]. Neither BPA nor E2 had any statistically
significant effect on Gh mRNA levels in males [Fig. 3(f);
F
(4, 25)
= 1.17; P= 0.35] or females [Fig. 3(g); F
(4, 25)
=
0.68; P= 0.61]. BPA and E2 also had no statistically
significant effects on Tshb mRNA levels in males
[Fig. 3(h); F
(4, 24)
= 0.92; P= 0.47] or females [Fig. 3(i);
F
(4, 24)
= 1.76; P= 0.17]. Finally, to determine whether the
decrease in Pit1 mRNA seen in males at PND7 would be
present after removal of BPA, we examined the 5-week-
old mice. No statistically significant effect of exposure
was found at 5 weeks in the males [Fig. 3(j); F
(4, 17)
= 0.28;
P= 0.89] or females [Fig. 3(k); F
(4, 18)
= 0.42; P= 0.79].
Experiment 1: BPA decreased Pomc mRNA in a
dose- and sex-specific manner
The last lineage examined was the TPIT lineage, which
includes the corticotrope and melanotrope cells. At
PND7, no statistically significant effect of BPA or E2
exposure was found on Tpit mRNA levels in males
[Fig. 4(a); F
(4, 25)
= 0.29; P= 0.88] or females [Fig. 4(b);
F
(4, 25)
= 0.74; P= 0.57]. A statistically significant effect of
exposure on Pomc mRNA levels was observed in males
[F
(4, 24)
= 4.01, P#0.01] and females [F
(4, 25)
= 3.31, P#
0.03]. The middle dose of BPA, BPA0.5, and E2 both
decreased Pomc mRNA in males [Fig. 4(c); P#0.02; P#
0.01]. In contrast, exposure to the lowest dose of BPA,
BPA0.05, and the highest dose, BPA50, showed no sta-
tistically significant change in Pomc mRNA [Fig. 4(c)]. In
females, Pomc mRNA was reduced with exposure to
Table 2. Five-Week Wet Weights and Measurements
Variable
Weight, g
Testes, %BW Ovaries, %BW Uteri, %BW Liver, %BW
AGD, mm/gBW
Male Female Male Female
Control 27.446 23.517 0.499 0.092 0.792 6.470 0.47 0.28
BPA0.05 26.950 22.350 0.506 0.112 0.937 6.570 0.50 0.29
BPA0.5 27.813 23.083 0.509 0.095 0.689 5.373 0.49 0.30
BPA50 29.743 23.136 0.505 0.110 0.760 6.309 0.51 0.31
E2 27.111 24.675 0.573 0.080 0.598 6.842 0.48 0.28
Abbreviations: AGD, anogenital distance; gBW, grams of body weight; %BW, percentage of body weight.
doi: 10.1210/en.2017-00565 https://academic.oup.com/endo 123
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
BPA0.5, BPA50, and E2 [Fig. 4(d); P#0.04, P#0.02,
and P#0.01, respectively]. No statistically significant
change in Pomc mRNA was observed with the BPA0.05
dose. However, POMC immunostaining and cortico-
trope number appeared unaffected by exposure in females
(Supplemental Fig. 1). Next, intrapituitary POMC
protein levels were examined by western blot analysis in
both males [Fig. 4(e)] and females [Fig. 4(f)], and no
statistically significant changes in protein levels were
detected in either sex. Next, Pomc mRNA levels were
Figure 1. Proliferation was not changed by neonatal exposure to BPA or E2. Proliferation was measured by Mki67 mRNA levels in (a) males and
(b) females at PND7, demonstrating no statistically significant change in proliferation. Also, at 5 weeks, no statistically significant change was
observed in proliferation in (c) males or (d) females (n = 3 to 6 for each sex). Circles represent individual mice, and average is represented by
a line. Proliferation, as assessed by phosphorylated histone H3 expression in sections of PND7 females, was similar among (e) control-, (f)
BPA0.05-, (g) BPA0.5-, and (h) BPA50-treated pituitary glands. Proliferation in male pituitary glands was also similar among (i) control, (j)
BPA0.05, (k) BPA0.5, and (l) BPA50 exposure (n = 3 to 4 for each sex and each condition).
124 Eckstrum et al Bisphenol A Effects on Postnatal Pituitary Gland Endocrinology, January 2018, 159(1):119131
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
assessed at 5 weeks. No statistically significant changes in
Pomc mRNA were observed in males [Fig. 4(g); F
(4, 17)
= 0.81;
P= 0.53]. In females, a statistically significant effect of
treatment was seen [F
(4, 17)
=4.18;P#0.02]. BPA0.5-
treated females had lower Pomc mRNA levels compared
with the levels in the control females [Fig. 4(h); P#0.03].
Those treated with BPA0.05 (P= 0.10), BPA50 (P= 0.08),
andE2(P= 0.09) had levels approaching statistical
significance.
Experiment 2: E2 and BPA decrease Pomc mRNA
directly at the pituitary
Owing to the striking suppression of Pomc mRNA in
vivo with both E2 and BPA, we sought to determine
whether these changes resulted from direct effects at the
level of the pituitary. PND1 pituitary glands were placed
in culture and treated with either E2 (10
28
M) or BPA
(4.4 310
25
M). In both males and females, E2 decreased
Pomc mRNA levels directly at the pituitary [Fig. 5(a)].
Next, pituitary glands were treated with BPA, which
substantially reduced Pomc mRNA in both sexes, similar
to the effects with E2 [Fig. 5(b)], suggesting these com-
pounds can act directly at the pituitary to exact changes in
Pomc expression similar to that found in vivo.
Experiment 2: Pomc mRNA is decreased by ESR1 and
ESR2 in females and by GPER in males
Because Pomc was suppressed by E2 and BPA directly
at the pituitary, the precise estrogen signaling pathway
that might be involved in this process was examined.
First, the pituitary glands were treated with an ESR1
selective agonist, PPT. PPT alone was unable to statis-
tically significantly suppress levels of Pomc mRNA [Fig.
6(a); male, P= 0.09; female, P= 0.17]. Next, pituitary
glands were treated with the ESR2 selective agonist,
DPN. DPN alone was also unable to statistically signif-
icantly suppress the levels of Pomc mRNA [Fig. 6(b);
male, P= 0.79; female, P= 0.93]. Because PPT or DPN
alone did not suppress Pomc sufficiently, the pituitary
glands were cotreated with PPT and DPN to determine
whether activation of both ESR1 and ESR2 would lead to
Figure 2. Gonadotrope markers were unchanged by neonatal BPA exposure, despite regulation by E2. Gonadotrope marker Nr5a1 mRNA was
measured in (a) males and (b) females at PND7, with no response to BPA. At 5 weeks, no effect of BPA or E2 was found in (c) males. (d)
However, suppression of Nr5a1 mRNA expression was found in females with exposure to BPA0.05 and E2. Lhb mRNA was decreased by E2 in (e)
males and (f) females at PND7 but (e) was unchanged by BPA. (g) At 5 weeks, Lhb was unaltered by any treatment group in males. (h) In females
at 5 weeks, E2 caused substantial repression of Lhb mRNA. For PND7, n = 5 to 6; for 5 weeks, n = 3 to 6. Circles represent individual mice, and
average is represented by a line.
#
P#0.05.
Table 3. Puberty Measurements
Variable Preputial Separation (d) Vaginal Opening (d)
Control 23.695 22.514
BPA0.05 23.700 23.533
BPA0.5 24.222 23.167
BPA50 24.104 23.422
E2 25.780 21.667
Day (d) of preputial separation and vaginal opening were monitored and
averaged for males (n = 3 to 5) and females (n = 4 to 6).
doi: 10.1210/en.2017-00565 https://academic.oup.com/endo 125
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
relevant repression. This cotreatment was able to sup-
press Pomc mRNA in females but not in males [Fig. 6(c);
male, P= 0.71; female, P#0.002].
To determine whether this regulation of Pomc might
occur through membrane estrogen receptors, pituitary
glands were treated with an estrogen dendrimer con-
jugate. This compound is too large to enter the nucleus
and thus only activates estrogen receptors in the plasma
membrane or cytoplasmic sites. Treatment of pituitary
glands with this compound did not lead to any statis-
tically significant changes in Pomc expression [Fig. 6(d);
male, P= 0.26; female, P= 0.91]. Next, cotreatment of
PPT, DPN, and estrogen dendrimer conjugate, meant to
activate both membrane and nuclear signaling path-
ways, showed suppression of Pomc in females but,
again, not in males [Fig. 6(e); male, P= 0.37; female, P#
0.003]. Finally, pituitary glands were treated with the
GPER selective agonist G1. G1 was able to decrease the
levels of Pomc mRNA in males but not in females [Fig.
6(f); male, P#0.01; female, P= 0.64]. This showed
that suppression of Pomc can occur through multi-
ple pathways and can be different between males
and females.
Discussion
We have demonstrated that critical windows of de-
velopment exist in which the mouse pituitary gland is
sensitive to BPA exposure and that differential effects
occur based on age and sex. Embryonic exposure to 0.5
or 50 mg/kg/d BPA increased pituitary proliferation and
gonadotrope number in a sex-specific manner (29).
Exposure to BPA during the neonatal period did not
affect proliferation and also had little effect on gona-
dotropes. However, we did see a decrease in Pit1
mRNA expression with lower doses of BPA in males
and sex-specific decreases in Pomc mRNA with ex-
posure to BPA and E2. Pomc mRNA regulation by BPA
and E2 can occur at the level of the pituitary in males
and females; however, sex-specific downregulation of
Pomc mRNA levels occurs with distinct estrogen re-
ceptor agonists. Therefore, these findings highlight that
the pituitary is dynamically affected by BPA exposure
in a sex-specific manner and emphasize the need for
examining different periods of exposure and times
after exposure.
Previously, our laboratory showed that embryonic
exposure to BPA in mice led to an increase in progenitor
proliferation and differentiation to the gonadotrope
Figure 3. BPA suppressed Pit1 mRNA at low doses in males but did
not alter transcription of PIT1 lineage hormones. Pit1 mRNA was
decreased at PND7 with 0.05 and 0.5 mg/kg/d BPA in (a) males but
not (b) females. (c) However, PIT1 protein was not changed by BPA
or E2 in males by western blot analysis. Prl mRNA was not altered
by BPA but was increased with E2 in (d) males and (e) females. No
difference was seen in Gh mRNA with any treatment in (f) males or
(g) females. Also, no difference in Tshb mRNA with BPA or E2
exposure in (h) males or (i) females was detected. Pit1 mRNA at
5 weeks was not changed in (j) males or (k) females. For quantitative
Figure 3. (Continued). PCR, n = 3 to 6; for PND7 western blot,
n = 3. Circles represent individual mice, and average is represented
by a line.
#
P#0.05.
126 Eckstrum et al Bisphenol A Effects on Postnatal Pituitary Gland Endocrinology, January 2018, 159(1):119131
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
lineage at PND0 in females (29). We found that the effect
of neonatal exposure to BPA was not similar to the effects
observed with embryonic dosing. Neonatal exposure
demonstrated no substantial change in proliferation or
gonadotropes in either sex. A higher concentration of
50 mg/kg/d BPA and direct stimulation of the neonatal
pituitary also did not regulate Lhb and Fshb mRNA (31),
demonstrating effectively that in the neonatal pituitary,
gonadotropes are less affected by BPA exposure than
during embryogenesis. Many reasons could exist for why
the neonatal period did not respond to BPA the same as
during the embryonic period. Pituitary cell composition
and receptor levels vary during the developmental period
(4042), which could alter the precise cellular dynamics
leading to these changes. Additionally, the functionality
of the HPG axis during these two periods might play a
role. Development of the HPG axis begins before birth (2)
and is of critical importance to the pituitary, because both
hpg (hypogonadal) mice lacking gonadotropin-releasing
hormone (GnRH) and kisspeptin receptor, GPR54,
knockout mice have lower serum gonadotropin levels at
birth (41, 43). However, after birth, the pituitary does not
respond to GnRH with increased luteinizing hormone
release until 2 weeks of age, despite a high frequency of
GnRH stimulation in the first week of life (44). This
suggests that the pituitary is hyporesponsive to stimuli
during the neonatal period, in contrast to the embryo or
the adult. Perhaps, late during the embryonic period, the
HPG axis and regulation of proliferation and Lhb mRNA
is active; however, after birth, the responsiveness of the
pituitary to GnRH and proliferation signals is decreased,
preventing disruption of Lhb mRNA and proliferation by
BPA during the neonatal period.
Despite an absence of similar changes in the neonatal
and embryonic periods, effects were observed that were
specific to neonatal exposure. The lowest doses of BPA,
within the range of human exposure, decreased Pit1
mRNA in males. To the best of our knowledge, BPA has
not been previously shown to affect Pit1 transcription.
This could be a nonestrogenic action because the
same effect was not seen with E2. However, E2 has
been shown to increase Pit1 mRNA in cell lines and
somatotrope-specific estrogen receptor-aknockout
mice in adults (45). It is possible that regulation of Pit1
mRNA by E2 was not seen in our experiments because
of the neonatal timing. BPA has been shown to bind
different receptors, including the androgen receptor,
estrogen-related receptor, thyroid hormone recep-
tor, peroxisome proliferator-activated receptor, aryl
hydrocarbon receptor, and glucocorticoid receptor
(46, 47). Therefore, it is possible that BPA might be
acting through one of these other receptors to reduce
Pit1 mRNA levels. Despite the decrease in Pit1 mRNA
with low doses of BPA in males, we did not see any effect
on the transcript levels of Prl,Gh,orTshb.Further-
more, we did not find any change in the protein levels of
PIT1, supporting the lack of changes in the transcrip-
tion of genes regulated by PIT1. These data suggest
the pituitary might use compensatory mechanisms to
Figure 4. Pomc mRNA was suppressed by BPA and E2 neonatal
exposure. Tpit mRNA was unchanged by BPA or E2 at PND7 in (a)
males and (b) females. Pomc mRNA was (c) decreased with 0.5 mg/kg/d
BPA and E2 at PND7 in males and (d) decreased with 0.5 mg/kg/d
BPA, 50 mg/kg/d BPA, and E2 in females. POMC protein was
unchanged by BPA in (e) males and (f) females. (g) Pomc mRNA was
not different with treatment at 5 weeks in males. (h) However,
a persistent decrease was seen in females treated with 0.5 mg/kg/d
BPA. For quantitative PCR, n = 3 to 6; for PND7 western blot, n = 3.
Circles represent individual mice, and average of combined sexes is
represented by a line.
#
P#0.05.
doi: 10.1210/en.2017-00565 https://academic.oup.com/endo 127
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
maintain consistent levels of PIT1 in the presence of
BPA exposure.
Neonatal BPA exposure also had striking effects on the
TPIT lineage that were not seen after embryonic dosing.
We observed a decrease in Pomc mRNA in the E2-,
BPA0.5-, and BPA50-exposed females and with BPA0.5
and E2 exposure in males, independent of the Tpit
mRNA changes. E2 exposure via an implanted 0.5-mg
pellet in the adult rat has been shown to reduce Pomc
mRNA in the pituitary, which also leads to decreased
plasma adrenocorticotropic hormone (ACTH) and cor-
ticosterone levels when stressed (48). Seeing the same
suppression of Pomc in the neonate suggests that the
mechanism by which this inhibition occurs is established
early on. However, the precise mechanism is unclear.
BPA50-exposed males did not show a statistically
significant decrease in Pomc mRNA. This represents a
crucial sex difference in Pomc mRNA regulation with
varying amounts of exposure and, potentially, a non-
monotonic dose response in males. Despite this, we did
not see a decrease in intrapituitary POMC protein ex-
pression or cell number. There could be a variety of
explanations, such as increased translation, to compen-
sate for decreased transcription. Alternatively, it could be
that hormone release is also affected,
and intrapituitary POMC levels are
kept consistent. Other studies in rats
have demonstrated the ability of 2 or
40 mg/kg/d BPA to regulate Pomc
mRNA levels and blood ACTH levels
in males compared with females in
response to stress (24, 49), emphasiz-
ing the importance of sex as a variable
in research on the HPA axis.
Of the parameters examined, few
statistically significant effects of BPA
exposure at 5 weeks after neonatal
dosing. A decrease was seen in Nr5a1
in BPA0.05-treated female pituitary
glands at 5 weeks; however, no cor-
responding decrease was seen in Lhb.
Additionally, changes in Pit1 at PND7
were not seen at 5 weeks. However, a
suppression of Pomc in BPA0.5-treated
females was found at 5 weeks, sug-
gesting the possibility of lasting effects
on the stress axis. Despite the relative
lack of changes due to BPA, E2 ex-
posure affected the reproductive axis
of 5-week females, leading to lower
Nr5a1 and Lhb levels and persistent
estrus. Therefore, neonatal E2 expo-
sure might have permanently altered
the HPG axis, which could affect fertility in females.
Although the effects of BPA were minimal on the 5-week
old pituitary gland, it is possible that changes could be
present at different time points. Additionally, these data
highlight that it is vital to examine exposure during
critical windows of development.
The strongest effect of neonatal dosing was a decrease
in Pomc mRNA with both E2 and BPA. Further analysis
demonstrated that this decrease might be a direct effect on
the pituitary itself. Although it has been reported that E2
can regulate Pomc mRNA, conflicting findings have been
reported on the direction and mechanism. In the adult rat
pituitary, E2 decreased Pomc mRNA, leading to de-
creased stress-related ACTH release (48). This effect,
however, might not be due to a direct effect on the pi-
tuitary gland because, in adult anterior pituitary-cultured
cells, E2 treatment did not alter Pomc mRNA expression.
However, in the neurointermediate lobe, E2 treatment
increased Pomc mRNA (50). In contrast, neonatal tes-
tosterone propionate decreased Pomc mRNA in the
adult, and treatment of E2 returned the levels of Pomc
mRNA to normal (51). Pomc is also expressed in the
hypothalamus, and E2 treatment decreases Pomc mRNA
in embryonic hypothalamic cultures (52). Alternatively,
Figure 5. Pomc mRNA was suppressed by BPA and E2 in pituitary cultures. Pomc mRNA was
suppressed by (a) 10
28
M estradiol and (b) 4.4 310
25
M BPA in pituitary explant cultures in
both males and females (E2 males, n = 6 to 10; E2 females, n = 10 to 15; BPA males, n = 9
to 15, BPA females, n = 7 to 14).
#
P#0.05.
128 Eckstrum et al Bisphenol A Effects on Postnatal Pituitary Gland Endocrinology, January 2018, 159(1):119131
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
in adult mice, E2 increased Pomc mRNA in POMC-
expressing neurons (53). Although different effects of
E2 on Pomc expression have been observed, it is clear
that E2 can regulate Pomc in an age-dependent and sex-
specific manner. The mechanism by which estradiol
regulates Pomc expression is not well understood;
however, in the hypothalamus ESR1 colocalizes with
POMC-expressing neurons (54), suggesting a role for
ESR1. In our studies of the neonatal pituitary gland,
ESR1 and ESR2, together, decreased the levels of Pomc
mRNA in females, but not in males. In contrast, acti-
vation of GPER decreased the levels of Pomc mRNA in
males but not in females. This interesting observation
can be explained by differing levels of estrogen recep-
tors or isoforms of the receptors. In the adult pituitary
gland, estrogen receptor mRNA isoforms are differen-
tially expressed in males and females. In addition, pitu-
itary expression of these isoforms can vary in females
depending onthe stage of the estrous cycle (55). Therefore,
it is possible that differences in receptor expression or
activation during this neonatal period could lead to the
differing pathways for Pomc mRNA downregulation and
should be further explored. However, it seems that
despite the possible differences in the roles of the various
estrogen receptors, it is clear that estrogen stimulation in
the neonatal pituitary leads to decreased Pomc mRNA
expression.
Conclusions
Overall, we found contrasting effects of BPA exposure in
the embryonic vs the neonatal period. This might be
demonstrating that the critical window for proliferation
and gonadotrope regulation is embryonic, and the critical
window for corticotrope regulation is neonatal. After
removal of BPA, minimal statistically significant effects
were seen, potentially demonstrating the plasticity of the
pituitary in regulating itself to maintain the proper cell
number and hormonal signaling. A commonality in both
the embryonic and the neonatal periods was the exis-
tence of sex-specific effects, which could hint at either
baseline differences in male and female pituitary glands
or differences in hormone signaling, both worthy of
further exploration. Finally, in our studies, we began to
explore the mechanism of estrogenic regulation of Pomc
mRNA. We observed that receptor-selective agonists
have sex-specific effects on Pomc gene expression. This
might suggest that BPA can activate different estrogen
receptors in males and females and, therefore, could
be a potential mechanism by which BPA mediates sex-
specific effects.
Acknowledgments
We thank Karen Weis and Liying Gao for technical assistance.
Financial Support: This work was supported by the Na-
tional Institutes of Health (Grant R01 DK076647 to L.T.R.;
Grant P01 ES022848 to J.A.F.; Grant DK015556 to J.A.K.; and
Grant T32 ES007326 to K.S.E. and W.W.). This work was also
supported by the Environmental Protection Agency (Grant RD-
83459301 to J.A.F.) and the Midwest Society of Toxicology
(Young Investigator Award to K.S.E.).
Correspondence: Lori T. Raetzman, PhD, Department
of Molecular and Integrative Physiology, University of Illi-
nois at Urbana-Champaign, 524 Burrill Hall, 407 South
Goodwin Avenue, Urbana, Illinois 61801. E-mail: raetzman@
life.illinois.edu.
Disclosure Summary: The authors have nothing to
disclose.
Figure 6. Pomc mRNA was regulated by various estrogen receptors in a sex-specific manner. (a) ESR1 selective agonist PPT and (b) ESR2 selective
agonist DPN did not affect Pomc mRNA. (c) PPT and DPN showed decreased Pomc mRNA in females. (d) Membrane estrogen signaling agonist,
estrogen dendrimer conjugate (ED), showed no regulation of Pomc mRNA. (e) PPT plus DPN plus ED decreased Pomc mRNA in females. (f) Finally,
the GPER agonist, G1, decreased Pomc mRNA in males. PPT males, n = 11; PPT females, n = 9 to 10; DPN males, n = 9 to 10; DPN females, n =
8; PPT plus DPN males, n = 11; PPT plus DPN females, n = 8 to 10; ED males, n = 10; ED females, n = 9; PPT plus DPN plus ED males, n = 7 to
13; PPT plus DPN plus ED females, n = 9 to 12; G1 males, n = 9 to 10; G1 females, n = 6 to 10.
#
P#0.05.
doi: 10.1210/en.2017-00565 https://academic.oup.com/endo 129
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
References
1. Davis SW, Ellsworth BS, Per´ez Millan MI, Gergics P, Schade V,
Foyouzi N, Brinkmeier ML, Mortensen AH, Camper SA. Pituitary
gland development and disease: from stem cell to hormone pro-
duction. Curr Top Dev Biol. 2013;106:147.
2. Pointis G, Mahoudeau JA. Release of immuno-reactive and bi-
ologically active LH from fetal mouse pituitary in response to
synthetic gonadotropin releasing factor (LRF). Experientia. 1976;
32(10):13471348.
3. Milkovi´c S, Milkovi´c K, Paunovc J. The initiation of fetal adre-
nocorticotrophic activity in the rat. Endocrinology. 1973;92(2):
380384.
4. Homma T, Sakakibara M, Yamada S, Kinoshita M, Iwata K,
Tomikawa J, Kanazawa T, Matsui H, Takatsu Y, Ohtaki T,
Matsumoto H, Uenoyama Y, Maeda K, Tsukamura H. Significance
of neonatal testicular sex steroids to defeminize anteroventral
periventricular kisspeptin neurons and the GnRH/LH surge system
in male rats. Biol Reprod. 2009;81(6):12161225.
5. McCormick CM, Furey BF, Child M, Sawyer MJ, Donohue SM.
Neonatal sex hormones have organizationaleffects on the
hypothalamic-pituitary-adrenal axis of male rats. Brain Res Dev
Brain Res. 1998;105(2):295307.
6. Shelby MD. NTP-CERHR monograph on the potential human
reproductive and developmental effects of bisphenol A. NTP
CERHR Mon. 2008;79(22):164 passim.
7. Herbst AL. The current status of the DES-exposed population.
Obstet Gynecol Annu. 1981;10:267278.
8. Markey CM, Michaelson CL, Veson EC, Sonnenschein C, Soto
AM. The mouse uterotrophic assay: a reevaluation of its validity in
assessing the estrogenicity of bisphenol A. Environ Health Perspect.
2001;109(1):5560.
9. Markey CM, Wadia PR, Rubin BS, Sonnenschein C, Soto AM.
Long-term effects of fetal exposure to low doses of the xenoestrogen
bisphenol-A in the female mouse genital tract. Biol Reprod. 2005;
72(6):13441351.
10. Vandenberg LN, Hauser R, Marcus M, Olea N, Welshons WV.
Human exposure to bisphenol A (BPA). Reprod Toxicol. 2007;
24(2):139177.
11. Nishihara E, Nagayama Y, Inoue S, Hiroi H, Muramatsu M,
Yamashita S, Koji T. Ontogenetic changes in the expression of
estrogen receptor alpha and beta in rat pituitary gland detected by
immunohistochemistry. Endocrinology. 2000;141(2):615620.
12. Ogasawara K, Nogami H, Tsuda MC, Gustafsson JA, Korach KS,
Ogawa S, Harigaya T, Hisano S. Hormonal regulation of prolactin
cell development in the fetal pituitary gland of the mouse. Endo-
crinology. 2009;150(2):10611068.
13. Hazell GG, Yao ST, Roper JA, Prossnitz ER, OCarroll AM, Lolait
SJ. Localisation of GPR30, a novel G protein-coupled oestrogen
receptor, suggests multiple functions in rodent brain and peripheral
tissues. J Endocrinol. 2009;202(2):223236.
14. Mitra SW, Hoskin E, Yudkovitz J, Pear L, Wilkinson HA, Hayashi
S, Pfaff DW, Ogawa S, Rohrer SP, Schaeffer JM, McEwen BS, Alves
SE. Immunolocalization of estrogen receptor beta in the mouse
brain: comparison with estrogen receptor alpha. Endocrinology.
2003;144(5):20552067.
15. Merchenthaler I, Lane MV, Numan S, Dellovade TL. Distribution
of estrogen receptor alpha and beta in the mouse central nervous
system: in vivo autoradiographic and immunocytochemical ana-
lyses. J Comp Neurol. 2004;473(2):270291.
16. Losa-Ward SM, Todd KL, McCaffrey KA, Tsutsui K, Patisaul HB.
Disrupted organization of RFamide pathways in the hypothalamus
is associated with advanced puberty in female rats neonatally ex-
posed to bisphenol A. Biol Reprod. 2012;87(2):28.
17. Monje L, Varayoud J, Mu~
noz-de-Toro M, Luque EH, Ramos JG.
Exposure of neonatal female rats to bisphenol A disrupts hypo-
thalamic LHRH pre-mRNA processing and estrogen receptor alpha
expression in nuclei controlling estrous cyclicity. Reprod Toxicol.
2010;30(4):625634.
18. Fern´andez M, Bianchi M, Lux-Lantos V, Libertun C. Neonatal
exposure to bisphenol a alters reproductive parameters and
Appendix. Antibody Table
Peptide/Protein
Target
Antigen
Sequence
(if Known) Name of Antibody
Manufacturer,
Catalog No.,
and/or Name of
Individual
Providing
the Antibody
Species Raised in;
Monoclonal or
Polyclonal
Dilution
Used RRID
ACTH Human ACTH
AA 1-24
Polyclonal rabbit
anti-human ACTH
DakoCytomation,
A0571
Rabbit; polyclonal 1:250 No longer
commercially
available
Mouse b-actin NA b-Actin (D6A8)
rabbit mAb
Cell Signaling
Technology,
8457
Rabbit; monoclonal 1:1000 AB_10950489
Chicken a-tubulin NA Mouse antia-tubulin
monoclonal antibody,
unconjugated,
clone DM1A
Sigma-Aldrich,
T9026
Mouse; monoclonal 1:5000 AB_477593
Rat PIT1 NA Anti-PIT1 Simon Rhodes Rabbit; polyclonal 1:1000 Not commercially
available
Rat bLH NA Anti-rbLH-IC National Hormone
and Peptide
Program,
Dr. A.F. Parlow
Rabbit; polyclonal 1:1000 AB_2665533
Human
phosphorylated
histone H3
Histone H3 at
serine 10
Anti-phosphorylated
histone H3 (Ser10)
antibody
Millipore Rabbit; polyclonal 1:1000 AB_310177
Abbreviations: AA, amino acid; AB, antibody; LHb, luteinizing hormone-b; mAb, monoclonal antibody; NA, not available; rbLH-IC, rat LH bfor
immunocytochemistry; RRID, Research Resource Identifier.
130 Eckstrum et al Bisphenol A Effects on Postnatal Pituitary Gland Endocrinology, January 2018, 159(1):119131
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
gonadotropin releasing hormone signaling in female rats. Environ
Health Perspect. 2009;117(5):757762.
19. Wang W, Hafner KS, Flaws JA. In utero bisphenol A exposure
disrupts germ cell nest breakdown and reduces fertility with age in
the mouse. Toxicol Appl Pharmacol. 2014;276(2):157164.
20. Howdeshell KL, Hotchkiss AK, Thayer KA, Vandenbergh JG, vom
Saal FS. Exposure to bisphenol A advances puberty. Nature. 1999;
401(6755):763764.
21. Khurana S, Ranmal S, Ben-Jonathan N. Exposure of newborn male
and female rats to environmental estrogens: delayed and sustained
hyperprolactinemia and alterations in estrogen receptor expression.
Endocrinology. 2000;141(12):45124517.
22. Ramirez MC, Bourguignon NS, Bonaventura MM, Lux-Lantos V,
Libertun C, Becu-Villalobos D. Neonatal xenoestrogen exposure
alters growth hormone-dependent liver proteins and genes in adult
female rats. Toxicol Lett. 2012;213(3):325331.
23. Zoeller RT, Bansal R, Parris C. Bisphenol-A, an environmental con-
taminant that acts as a thyroid hormone receptor antagonist in vitro,
increases serum thyroxine, and alters RC3/neurogranin expression in
the developing rat brain. Endocrinology. 2005;146(2):607612.
24. Chen F, Zhou L, Bai Y, Zhou R, Chen L. Sex differences in the adult
HPA axis and affective behaviors are altered by perinatal exposure
to a low dose of bisphenol A. Brain Res. 2014;1571:1224.
25. Patchev VK, Hayashi S, Orikasa C, Almeida OF. Implications of
estrogen-dependent brain organization for gender differences in
hypothalamo-pituitary-adrenal regulation. FASEB J. 1995;9(5):
419423.
26. Rubin BS, Lenkowski JR, Schaeberle CM, Vandenberg LN,
Ronsheim PM, Soto AM. Evidence of altered brain sexual
differentiation in mice exposed perinatally to low, environmen-
tally relevant levels of bisphenol A. Endocrinology. 2006;147(8):
36813691.
27. Cao J, Mickens JA, McCaffrey KA, Leyrer SM, Patisaul HB.
Neonatal Bisphenol A exposure alters sexually dimorphic gene
expression in the postnatal rat hypothalamus. Neurotoxicology.
2012;33(1):2336.
28. Stahlhut RW, Welshons WV, Swan SH. Bisphenol A data in
NHANES suggest longer than expected half-life, substantial non-
food exposure, or both. Environ Health Perspect. 2009;117(5):
784789.
29. Brannick KE, Craig ZR, Himes AD, Peretz JR, Wang W, Flaws JA,
Raetzman LT. Prenatal exposure to low doses of bisphenol A in-
creases pituitary proliferation and gonadotroph number in female
mice offspring at birth. Biol Reprod. 2012;87(4):82.
30. Wu YJ, Chen DW, Liu JL, Zhang JH, Luo HS, Cui S. Estradiol
promotes pituitary cell proliferation and gonadotroph differenti-
ation at different doses and with different mechanisms in chick
embryo. Steroids. 2009;74(4-5):441448.
31. Eckstrum KS, Weis KE, Baur NG, Yoshihara Y, Raetzman LT.
Icam5 expression exhibits sex differences in the neonatal pituitary
and is regulated by estradiol and bisphenol A. Endocrinology.
2016;157(4):14081420.
32. Lakind JS, Naiman DQ. Bisphenol A (BPA) daily intakes in the
United States: estimates from the 2003-2004 NHANES urinary
BPA data. J Expo Sci Environ Epidemiol. 2008;18(6):608615.
33. US Environmental Protection Agency. Health assessment in-
formation on bisphenol A (CASRN 80-05-7). Available at:http://
cfpub.epa.gov/ncea/iris/index.cfm?fuseaction=iris.showQuickview&
substance_nmbr=0356#reforal. First online 1998. Accessed 24
August 2017.
34. Gremeaux L, Fu Q, Chen J, Vankelecom H. Activated phenotype of the
pituitary stem/progenitor cell compartment during the early-postnatal
maturation phase of the gland. Stem Cells Dev. 2012;21(5):801813.
35. Caligioni CS. Assessing reproductive status/stages in mice. Curr
Protoc Neurosci. 2009;Appendix 4:Appendix 4I.
36. Harrington WR, Kim SH, Funk CC, Madak-Erdogan Z, Schiff R,
Katzenellenbogen JA, Katzenellenbogen BS. Estrogen dendrimer
conjugates that preferentially activate extranuclear, nongenomic
versus genomic pathways of estrogen action. Mol Endocrinol.
2006;20(3):491502.
37. Kim SH, Katzenellenbogen JA. Hormone-PAMAM dendrimer
conjugates: polymer dynamics and tether structure affect ligand
access to receptors. Angew Chem Int Ed Engl. 2006;45(43):
72437248.
38. Nantie LB, Himes AD, Getz DR, Raetzman LT. Notch signaling in
postnatal pituitary expansion: proliferation, progenitors, and cell
specification. Mol Endocrinol. 2014;28(5):731744.
39. Goldberg LB, Aujla PK, Raetzman LT. Persistent expression of
activated Notch inhibits corticotrope and melanotrope differenti-
ation and results in dysfunction of the HPA axis. Dev Biol. 2011;
358(1):2332.
40. Yoshimura F, Harumiya K, Kiyama H. Light and electron mi-
croscopic studies of the cytogenesis of anterior pituitary cells in
perinatal rats in reference to the development of target organs. Arch
Histol Jpn. 1970;31(3):333369.
41. Carbajo-P´erez E, Watanabe YG. Cellular proliferation in the an-
terior pituitary of the rat during the postnatal period. Cell Tissue
Res. 1990;261(2):333338.
42. Pasqualini C, Guivarch D, Boxberg YV, Nothias F, Vincent JD,
Vernier P. Stage- and region-specific expression of estrogen receptor
aisoforms during ontogeny of the pituitary gland. Endocrinology.
1999;140(6):27812789.
43. Poling MC, Kauffman AS. Sexually dimorphic testosterone secretion
in prenatal and neonatal mice is independent of kisspeptin-Kiss1r
and GnRH signaling. Endocrinology. 2012;153(2):782793.
44. Glanowska KM, Burger LL, Moenter SM. Development of
gonadotropin-releasing hormone secretion and pituitary response.
J Neurosci. 2014;34(45):1506015069.
45. Avtanski D, Novaira HJ, Wu S, Romero CJ, Kineman R, Luque
RM, Wondisford F, Radovick S. Both estrogen receptor aand b
stimulate pituitary GH gene expression. Mol Endocrinol. 2014;
28(1):4052.
46. Acconcia F, Pallottini V, Marino M. Molecular mechanisms of
action of BPA. Dose Response. 2015;13(4):1559325815610582.
47. Prasanth GK, Divya LM, Sadasivan C. Bisphenol-A can bind to
human glucocorticoid receptor as an agonist: an in silico study.
J Appl Toxicol. 2010;30(8):769774.
48. Redei E, Li L, Halasz I, McGivern RF, Aird F. Fast glucocorticoid
feedback inhibition of ACTH secretion in the ovariectomized rat:
effect of chronic estrogen and progesterone. Neuroendocrinology.
1994;60(2):113123.
49. Panagiotidou E, Zerva S, Mitsiou DJ, Alexis MN, Kitraki E.
Perinatal exposure to low-dose bisphenol A affects the neuro-
endocrine stress response in rats. J Endocrinol. 2014;220(3):
207218.
50. Matsumura R, Takeuchi S, Takahashi S. Effect of estrogen on
melanocortin-3 receptor mRNA expression in mouse pituitary
glands in vivo and in vitro. Neuroendocrinology. 2004;80(3):
143151.
51. Seale JV, Wood SA, Atkinson HC, Harbuz MS, Lightman SL.
Postnatal masculinization alters the HPA axis phenotype in the
adult female rat. J Physiol. 2005;563(Pt 1):265274.
52. Gao Y, He JR, Kapcala LP. Estrogen inhibits hypothalamic pro-
opiomelanocortin gene expression in hypothalamic neuronal cul-
tures. Brain Res Dev Brain Res. 1997;45(2):340344.
53. PelletierG,LiS,Luu-TheV,LabrieF.Oestrogenicregulationofpro-
opiomelanocortin, neuropeptide Y and corticotrophin-releasing hormone
mRNAs in mouse hypothalamus. J Neuroendocrinol.2007;19(6):
426431.
54. de Souza FSJ, Nasif S, L ´opez-Leal R, Levi DH, Low MJ, Rubinsten M.
The estrogen receptor acolocalizes with proopiomelanocortin in hy-
pothalamic neurons and bin ds to a conserved motif present in the neuron-
specific enhancer nPE2. Eur J Pharmacol. 2011;660(1):181187.
55. Demay F, Tiffoche C, Thieulant M-L. Sex- and cell-specific ex-
pression of an estrogen receptor isoform in the pituitary gland.
Neuroendocrinology. 1996;63(6):522529.
doi: 10.1210/en.2017-00565 https://academic.oup.com/endo 131
Downloaded from https://academic.oup.com/endo/article-abstract/159/1/119/4349657 by guest on 19 May 2020
... Critical periods of murine pituitary gland development which consist of the (i) formation of embryonic gland and (ii) the enlargement of neonatal hormone cell are particularly vulnerable to the adverse effects of EDCs, in which the female number of gonadotroph and proliferation was upregulated at both 0.5 and 50 μg/kg/day BPA during the embryonic development [47]. In contrast, Eckstrum et al. [48] have revealed that neonatal BPA exposure alters sex-specific expressions of gene which are different compared to the observations for prenatal exposure. Pituitary Pit1 mRNA were found to be downregulated in male only (0.05 and 0.5 μg/kg/day BPA), while Pomc mRNA levels were decreased in both male (0.5 μg/kg/d BPA) and female (0.5 and 50 μg/kg/day BPA) murines at PND7, with no alterations in the protein expression. ...
... The usage of ER subtype-selective agonists on pituitary organ cultures revealed that proopiomelanocortin (Pomc) mRNA suppression by low-dose BPA was influenced by the male murine G-protein-coupled ER and female murine ERα and ERβ, which meant that different mechanisms underlay the sexually dimorphic repression of Pomc caused by BPA exposure [48]. Other than that, zebrafish oocyte maturation was inhibited by 0.1 μM BPA via the nongenomic oestrogenic pathways involving GPER (with high binding affinity) and EGFR [23]. ...
Article
Full-text available
For the past two decades, growing research has been pointing to multiple repercussions of bisphenol A (BPA) exposure to human health. BPA is a synthetic oestrogen which primarily targets the endocrine system; however, the compound also disturbs other systemic organ functions, in which the magnitude of impacts in those other systems is as comparable to those in the endocrine system. To date, the discoveries on the association between BPA and health outcomes mainly came from animal and in vitro studies, with limited human studies which emphasised on children’s health. In this comprehensive review, we summarised studies on human, in vivo and in vitro models to understand the consequences of pre-, post- and perinatal BPA exposure on the perinatal, children and adult health, encompassing cardiovascular, neurodevelopmental, endocrine and reproductive effects. Conclusion: Evidence from in vitro and animal studies may provide further support and better understanding on the correlation between environmental BPA exposure and its detrimental effects in humans and child development, despite the difficulties to draw direct causal relations of BPA effects on the pathophysiology of the diseases/syndromes in children, due to differences in body system complexity between children and adults, as well as between animal and in vitro models and humans. What is known: • Very limited reviews are available on how BPA adversely affects children’s health. • Previous papers mainly covered two systems in children. What is new: • Comprehensive review on the detrimental effects of BPA on children health outcomes, including expectations on adult health outcomes following perinatal BPA exposure, as well as covering a small part of BPA alternatives. • Essentially, BPA exposure during pregnancy has huge impacts on the foetus in which it may cause changes in foetal epigenetic programming, resulting in disease onsets during childhood as well as adulthood.
... Особую чувствительность к повреждающему действию ЭД проявляют эндокринные органы плодов и новорожденных. Помимо канцерогенного действия при этом может изменяться половая диморфная организация гипоталамуса с последующим нарушением репродуктивной функции у взрослых [23][24][25]. ...
Article
The main aim of the study is to analyze modern knowledge on endocrine disruptors, non-genotoxic carcinogens, contributing significantly to the total level of contamination of the biosphere by anthropogenic blastomogens. Material and Methods . For the review preparation, we analyzed articles on molecular mechanisms of the effects of endocrine disruptors, available at biomedical literature databases sciVerse scopus, pubmed, Web of science, Rsci. The review cited 65 recent publications, 21 of them being published over the past three years, 3 papers being the official documents on hazards associated with the use of endocrine disruptors, and 10 papers presenting a background to separate endocrine disruptors into the group of compounds with specific functional activity. Results. The role of endocrine disruptors involves the development of the tumors of reproductive organs. They may reveal the properties of strong agonists or antagonists disrupting the hormonal balance by inhibition of the synthesis of natural hormones, their secretion, transport, metabolism, binding or degradation. In addition, they activate alternative proliferation signaling pathways by activating g-proteincoupled receptors, estrogen-bound eRRγ or/and β-adrenergic receptors. The main mechanism of action of endocrine disruptors is the induction of epigenetic modifications, in particular, methylation/demethylation of cpg dNa islands, histone modifications and changes in expression of non-coding RNa. since the effects of endocrine disruptors are nonlinear, they can be caused by concentrations corresponding to the real content in the biosphere and detected in the umbilical blood and breast milk. In addition, they contribute to the metabolic activation of the procarcinogens, the main component of air pollution, by activation of the cytochrome p450 isoforms. Conclusion . Endocrine disruptors can underlie the development of tumors of hormone-dependent organs both at direct and transplacental exposure.
... More recently, several authors reported that exposure of neonatal mice to BPA can disrupt pituitary development, with critical effects on their pubertal and hence gonadal development. The described effects comprise a measurable downregulation of the mRNA expression of several genes (including pomc and Icam5) involving different estrogen-related pathways, with sex-specific differences [68,69]. Furthermore, there is evidence from an animal study that the spectrum of reproductive alterations induced by chronic BPA exposure includes reproductive hormone alterations (increased gonadotropins and estradiol, reduced testosterone), spermatogenesis hindrance and increased oxidative stress [70]. ...
Article
Full-text available
Great attention has been paid in recent years to the harmful effects of various chemicals that interfere with our natural hormone balance, collectively known as endocrine-disrupting chemicals (EDCs) or endocrine disruptors. The effects on the reproductive system of bisphenol A (BPA) and phthalates have received particular attention: while they have a short half-life, they are so widespread that human exposure can be considered as continuous. Evidence is often limited to the animal model, disregarding the likelihood of human exposure to a mixture of contaminants. Data from animal models show that maternal exposure probably has harmful effects on the male fetus, with an increased risk of urogenital developmental abnormalities. After birth, exposure is associated with changes in the hypothalamic-pituitary-testicular axis, hindering the development and function of the male genital pathways through the mediation of inflammatory mechanisms and oxidative stress. The epidemiological and clinical evidence, while generally confirming the association between reproductive abnormalities and some phthalate esters and BPA, is more contradictory, with wildly different findings. The aim of this review is therefore to provide an update of the potential mechanisms of the damage caused by BPA and phthalates to reproductive function and a review of the clinical evidence currently available in the literature.
Article
Background: The Consortium Linking Academic and Regulatory Insights on Bisphenol A Toxicity (CLARITY-BPA) was a collaborative research effort to better link academic research with governmental guideline studies. This review explores the secondary goal of CLARITY-BPA: to identify endpoints or technologies from CLARITY-BPA and prior/concurrent literature from these laboratories that may enhance the capacity of rodent toxicity studies to detect endocrine disrupting chemicals (EDCs). Methods: A systematic literature search was conducted with search terms for BPA and the CLARITY-BPA participants. Relevant studies employed a laboratory rodent model and reported results on 1 of the 10 organs/organ systems evaluated in CLARITY-BPA (brain and behavior, cardiac, immune, mammary gland, ovary, penile function, prostate gland and urethra, testis and epididymis, thyroid hormone and metabolism, and uterus). Study design and findings were summarized, and a risk-of-bias assessment was conducted. Results: Several endpoints and methods were identified as potentially helpful to detect effects of EDCs. For example, molecular and quantitative morphological approaches were sensitive in detecting alterations in early postnatal development of the brain, ovary, and mammary glands. Hormone challenge studies mimicking human aging reported increased susceptibility of the prostate to disease following developmental BPA exposure. Statistical analyses for nonmonotonic dose responses, and computational approaches assessing multiple treatment-related outcomes concurrently in linked hormone-sensitive organ systems, reported effects at low BPA doses. Conclusions: This review provided an opportunity to evaluate the unique insights provided by nontraditional assessments in CLARITY-BPA to identify technologies and endpoints to enhance detection of EDCs in future studies.
Chapter
This chapter provides a practical overview of reproductive and developmental toxicology, with a focus on considerations for human health risk assessment. It provides a brief overview of normal reproduction and development, as well as examples of how toxic agents may impact these processes. Default assumptions for risk assessment of reproductive and developmental toxicants are discussed, and the types of endpoints typically evaluated in animal toxicology studies and epidemiology studies are summarized. A brief overview is also provided of other data that can be useful for hazard identification of reproductive and developmental toxicants, including mechanistic and new approach methodologies, pharmacokinetics/pharmacodynamics, and considerations for the risk assessment of mixtures. Testing procedures and guidelines for reproductive and developmental toxicants (including pharmaceuticals and industrial or environmental chemicals) are summarized.
Article
Polychlorinated-biphenyls (PCBs) are industrial compounds, which were widely used in manufacturing of electrical parts and transformers. Despite being banned in 1979 due to human health concerns, they persist in the environment. In humans and experimental model systems, PCBs elicit toxicity in part by acting as endocrine-disrupting chemicals (EDCs). Aroclor 1221 (A1221) is a weakly estrogenic PCB mixture known to alter reproductive function in rodents. EDCs can impact hormone signaling at any level of the hypothalamic-pituitary-gonadal (HPG) axis, and we investigated the effects of A1221 exposure during the prenatal and postnatal developmental periods on pituitary hormone and steroid receptor expression in female rats. Examining offspring at 3 ages, postnatal day 8 (P8), P32 and P60, we found that prenatal exposure to A1221 increased P8 neonate pituitary luteinizing hormone beta (Lhb) mRNA and LHβ gonadotrope cell number while decreasing LH serum hormone concentration. No changes in pituitary hormone or hormone receptor gene expression were observed peri-puberty at P32. In reproductively mature rats at P60, we found pituitary follicle stimulating hormone beta (Fshb) mRNA levels increased by prenatal A1221 exposure with no corresponding alterations in FSH hormone or FSHβ expressing cell number. Estrogen receptor alpha (ERα) mRNA and protein levels were also increased at P60, but only following postnatal A1221 dosing. Together, these data illustrate that exposure to the PCB A1221, during critical developmental windows, alters pituitary gonadotropin hormone subunits and ERα levels in offspring at different phases of maturation, potentially impacting reproductive function in concert with other components of the HPG axis.
Article
Full-text available
In 2015, EFSA established a temporary tolerable daily intake (t-TDI) for BPA of 4 μg/kg body weight (bw) per day. In 2016, the European Commission mandated EFSA to re-evaluate the risks to public health from the presence of BPA in foodstuffs and to establish a tolerable daily intake (TDI). For this re-evaluation, a pre-established protocol was used that had undergone public consultation. The CEP Panel concluded that it is Unlikely to Very Unlikely that BPA presents a genotoxic hazard through a direct mechanism. Taking into consideration the evidence from animal data and support from human observational studies, the immune system was identified as most sensitive to BPA exposure. An effect on Th17 cells in mice was identified as the critical effect; these cells are pivotal in cellular immune mechanisms and involved in the development of inflammatory conditions, including autoimmunity and lung inflammation. A reference point (RP) of 8.2 ng/kg bw per day, expressed as human equivalent dose, was identified for the critical effect. Uncertainty analysis assessed a probability of 57-73% that the lowest estimated Benchmark Dose (BMD) for other health effects was below the RP based on Th17 cells. In view of this, the CEP Panel judged that an additional uncertainty factor (UF) of 2 was needed for establishing the TDI. Applying an overall UF of 50 to the RP, a TDI of 0.2 ng BPA/kg bw per day was established. Comparison of this TDI with the dietary exposure estimates from the 2015 EFSA opinion showed that both the mean and the 95th percentile dietary exposures in all age groups exceeded the TDI by two to three orders of magnitude. Even considering the uncertainty in the exposure assessment, the exceedance being so large, the CEP Panel concluded that there is a health concern from dietary BPA exposure.
Article
Tris (1,3-dichloro-2-propyl) phosphate (TDCIPP), an alternative to brominated flame retardants, might pose an exposure risk to humans and wild animals during fetal development. Our recent study suggested that short-term TDCIPP exposure during early development caused sex-dependent behavioral alteration in adults. In the present study, multigenerational neurodevelopmental toxicity upon early-life exposure of parental zebrafish was evaluated, and the possible underlying mechanisms were further explored. Specifically, after embryonic exposure (0–10 days post-fertilization, dpf) to TDCIPP (0, 0.01, 0.10, and 1.00 μM), zebrafish larvae were cultured in clean water until the sexually matured to produce progeny (F1). The results confirmed neurodevelopmental toxicity in F1 larvae characterized by changes of developmental endpoints, reduced thigmotaxis, as well as altered transcription of genes including myelin basic protein a (mbpa), growth associated protein (gap43) and synapsin IIa (syn2a). Sex-specific changes in thyroid hormones (THs) indicated the relationship of abnormal THs levels with previously reported neurotoxicity in adult females after early-life exposure to TDCIPP. Similar changing profiles of TH levels (increased T3 and decreased T4) in adult females and F1 eggs, but not in F1 larvae, suggested that the TH disruptions were primarily inherited from the maternal fish. Further results demonstrated hypermethylation of global DNA and key genes related to TH transport including transthyretin (ttr) and solute carrier family 16 member 2 (slc16a2), which might affect the transport of THs to target tissues, thus at least partially contributing to the neurodevelopmental toxicity in F1 larvae. Overall, our results confirmed that early-life TDCIPP exposure of parental fish could affect the early neurodevelopment of F1 offspring. The underlying mechanism could involve altered TH levels inherited from maternal zebrafish and epigenetic modifications in F1 larvae.
Chapter
Consistent evidence indicates that endocrine disrupting chemicals (EDCs) often induce different adverse effects in males and females, in laboratory animals as well as in humans. This phenomenon is, at least partly, a consequence of sex-specific levels of steroid hormones and/or steroid hormone receptors in tissues and organisms, which also vary throughout development before full sexual maturity. This implies the need for sex-specific evaluation when assessing whether and through what mechanism(s) a substance has an endocrine disrupting effect. Investigations of EDCs and their mixtures on animal models may be of great significance, yet interpretation needs specific care as, for example, rodents show differences from humans with regard to the homeostasis of steroid hormones as well as hormone receptors. It is of the utmost interest to explore sex-related differences in EDCs’ effects on non-reproductive tissues: in particular, this chapter discusses the research findings concerning the immune system, adipose tissue and especially neurobehavioral development. The assessment of EDC effects in mechanistic, toxicological or epidemiological studies should always include an analysis of sex-related differences.
Article
Full-text available
Bisphenol A (BPA) exposure has been associated with serious endocrine-disrupting effects in humans and wildlife. Toxicological and epidemiological studies evidenced that BPA increases body mass index and disrupts normal cardiovascular physiology by interfering with endogenous hormones in rodents, nonhuman primates, and cell culture test systems. The BPA concentration derived from these experiments were used by government regulatory agencies to determine the safe exposure levels of BPA in humans. However, accumulating literature in vivo and in vitro indicate that at concentrations lower than that reported in toxicological studies, BPA could elicit a different endocrine-disrupting capacity. To further complicate this picture, BPA effects rely on several and diverse mechanisms that converge upon endocrine and reproductive systems. If all or just few of these mechanisms concur to the endocrine-disrupting potential of low doses of BPA is at present still unclear. Thus, taking into account that the incidence and/or prevalence of health problems associated with endocrine disruption have increased worldwide, the goal of the present review is to give an overview of the many mechanisms of BPA action in order to decipher whether different mechanisms are at the root of the effect of low dose of BPA on endocrine system.
Article
Full-text available
Acquisition of a mature pattern of gonadotropin-releasing hormone (GnRH) secretion from the CNS is a hallmark of the pubertal process. Little is known about GnRH release during sexual maturation, but it is assumed to be minimal before later stages of puberty. We studied spontaneous GnRH secretion in brain slices from male mice during perinatal and postnatal development using fast-scan cyclic voltammetry (FSCV) to detect directly the oxidation of secreted GnRH. There was good correspondence between the frequency of GnRH release detected by FSCV in the median eminence of slices from adults with previous reports of in vivo luteinizing hormone (LH) pulse frequency. The frequency of GnRH release in the late embryonic stage was surprisingly high, reaching a maximum in newborns and remaining elevated in 1-week-old animals despite low LH levels. Early high-frequency GnRH release was similar in wild-type and kisspeptin knock-out mice indicating that this release is independent of kisspeptin-mediated excitation. In vivo treatment with testosterone or in vitro treatment with gonadotropin-inhibitory hormone (GnIH) reduced GnRH release frequency in slices from 1-week-old mice. RF9, a putative GnIH antagonist, restored GnRH release in slices from testosterone-treated mice, suggesting that testosterone inhibition may be GnIH-dependent. At 2-3 weeks, GnRH release is suppressed before attaining adult patterns. Reduction in early life spontaneous GnRH release frequency coincides with the onset of the ability of exogenous GnRH to induce pituitary LH secretion. These findings suggest that lack of pituitary secretory response, not lack of GnRH release, initially blocks downstream activation of the reproductive system.
Article
Full-text available
Bisphenol A (BPA) is an estrogen-mimicking endocrine disruptor. Early life exposures to low BPA doses exert long lasting effects on animals' reproductive and brain physiology. However little is known about BPA effects on the stress response system. Given the interaction of sex and stress hormones, we examined the effect of a low perinatal BPA exposure on the function of the hypothalamic-pituitary-adrenal (HPA) axis at rest and upon application of acute stress. Throughout pregnancy and lactation rats received daily 40 microgram BPA/kg body weight orally via cornflakes. We studied the effect of this low but chronic exposure to BPA in the male and female offspring at puberty. BPA exposure led to abnormal adrenal histology including reduced zona reticularis especially in male offspring, hyperplasia of zona fasciculata in both sexes and increased adrenal weight in BPA females. BPA-treated females had increased basal corticosterone and reduced hypothalamic glucocorticoid receptors (GR) levels. Stressed BPA females exhibited an anxious-like behavioral coping, a less rigorous corticosterone response and did not down-regulate GR in the hypothalamus, compared to control females. BPA males exhibited a heightened corticosterone stress response compared to females; they also displayed increased pro-opiomelanocortin mRNA levels and retained the pre stress levels of pituitary CRH-receptor1, compared to control males. We found that perinatal chronic exposure to a low dose of BPA perturbs the basal and stress-induced activity of the HPA axis in a sexually dimorphic manner at adolescence. Exposure to BPA might contribute to increased susceptibility to stress-related disorders in later life.
Article
The expression time course of estrogen receptor α (ERα) was analyzed by RT-PCR in fetal and newborn rat pituitaries. In addition to the classical ERα messenger RNA (mRNA), three shorter transcripts were detected and subsequently cloned. Sequence analysis showed that they corresponded to ERα mRNAs lacking exon 3 (which encodes a zinc finger in the DNA-binding domain), exon 4 (which encodes the nuclear localization signal and part of the steroid-binding domain), or both exons 3 and 4. As analyzed by RT-PCR and ribonuclease protection assay, the respective expression levels of the different transcripts varied dramatically during pituitary development; short forms appeared 4 days before full-length ERα mRNA. On Western blots from rat pituitaries of different ages, an ERα-specific antiserum labeled four protein bands of the expected molecular weights, revealing that all four ERα mRNAs are translated in vivo. Immunocytochemistry, using the same antiserum, showed the ERα to be present first in the cytosol of int...
Article
Endocrine disrupting chemicals (EDCs) are prevalent in the environment and can impair reproductive success by affecting the hypothalamic-pituitary-gonadal (HPG) axis. The developing pituitary gland is sensitive to exposure to EDCs, such as bisphenol A (BPA), and sex-specific effects can occur. However, effects on the critical window of neonatal pituitary gland development in mice have not been explored. Therefore, this study determined baseline gene expression in male and female pituitaries and consequences of environmental exposure to 17β-estradiol (E2) and BPA on transcription of genes exhibiting sex differences during the neonatal period. Through microarray and qRT-PCR analysis of pituitaries at PND1, three genes were differentially expressed between males and females: Lhb, Fshb, and intracellular adhesion molecule-5 (Icam5). To see if E2 and BPA exposure regulates these genes, pituitaries were cultured at postnatal day (PND)1 with 10(-8)M E2 or 4.4x10(-6)M BPA. E2 decreased expression of Lhb, Fshb, and Icam5 mRNA in females, but only significantly decreased expression of Icam5 in males. BPA decreased expression of Icam5 similarly to E2, but it did not affect Lhb or Fshb. Importantly, in vivo exposure to 50 μ g/kg/day E2 from PND0-7 decreased expression of Lhb, Fshb, and Icam5 mRNA in both males and females while 50 mg/kg/day BPA exposure during the same time frame decreased expression of Icam5 in females only. Overall, we have uncovered that genes differentially expressed between the sexes can be regulated in part by hormonal and chemical signals in vivo and directly at the pituitary and can be regulated in a sex-specific manner.
Article
The National Toxicology Program (NTP) Center for the Evaluation of Risks to Human Reproduction (CERHR) conducted an evaluation of the potential for bisphenol A to cause adverse effects on reproduction and development in humans. The CERHR Expert Panel on Bisphenol A completed its evaluation in August 2007. CERHR selected bisphenol A for evaluation because of the: widespread human exposure; public concern for possible health effects from human exposures; high production volume; evidence of reproductive and developmental toxicity in laboratory animal studies Bisphenol A (CAS RN: 80-05-7) is a high production volume chemical used primarily in the production of polycarbonate plastics and epoxy resins. Polycarbonate plastics are used in some food and drink containers; the resins are used as lacquers to coat metal products such as food cans, bottle tops, and water supply pipes. To a lesser extent bisphenol A is used in the production of polyester resins, polysulfone resins, polyacrylate resins, and flame retardants. In addition, bisphenol A is used in the processing of polyvinyl chloride plastic and in the recycling of thermal paper. Some polymers used in dental sealants and tooth coatings contain bisphenol A. The primary source of exposure to bisphenol A for most people is assumed to occur through the diet. While air, dust, and water (including skin contact during bathing and swimming) are other possible sources of exposure, bisphenol A in food and beverages accounts for the majority of daily human exposure. The highest estimated daily intakes of bisphenol A in the general population occur in infants and children. The results of this bisphenol A evaluation are published in an NTP-CERHR Monograph that includes the (1) NTP Brief and (2) Expert Panel Report on the Reproductive and Developmental Toxicity of Bisphenol A. Additional information related to the evaluation process, including the peer review report for the NTP Brief and public comments received on the draft NTP Brief and the final expert panel report, are available on the CERHR website (http://cerhr.niehs.nih.gov/). See bisphenol A under "CERHR Chemicals" on the homepage or go directly to http://cerhr.niehs. nih.gov/chemicals/bisphenol/bisphenol.html). The NTP reached the following conclusions on the possible effects of exposure to bisphenol A on human development and reproduction. Note that the possible levels of concern, from lowest to highest, are negligible concern, minimal concern, some concern, concern, and serious concern. The NTP has some concern for effects on the brain, behavior, and prostate gland in fetuses, infants, and children at current human exposures to bisphenol A. The NTP has minimal concern for effects on the mammary gland and an earlier age for puberty for females in fetuses, infants, and children at current human exposures to bisphenol A. The NTP has negligible concern that exposure of pregnant women to bisphenol A will result in fetal or neonatal mortality, birth defects, or reduced birth weight and growth in their offspring. The NTP has negligible concern that exposure to bisphenol A will cause reproductive effects in non-occupationally exposed adults and minimal concern for workers exposed to higher levels in occupational settings. NTP will transmit the NTP-CERHR Monograph on Bisphenol A to federal and state agencies, interested parties, and the public and make it available in electronic PDF format on the CERHR web site (http://cerhr.niehs.nih.gov) and in printed text or CD from CERHR.
Article
Bisphenol A (BPA), an estrogen-mimicking endocrine disrupter, when administered perinatally can affect affective behaviors in adult rodents, however the underlying mechanisms remain largely unclear. Postnatal day (PND) 80 vehicle-injected control female rats showed more obvious depression- and anxiety-like behaviors than males, indicative of sexually dimorphic affective behaviors. When female breeders were subcutaneously injected with BPA (2µg/kg) from gestation day 10 to lactation day 7, sex difference of affective behaviors was impaired in their offspring (PND80 BPA-rats), as results that female BPA-rats showed a visible "antianxiety-like" behavior, and male BPA-rats increased depression-like behavior compared to vehicle-injected controls. Notably, basal levels of serum corticosterone and adrenocorticotropin (ACTH), and corticotropin-releasing hormone mRNA were increased in male BPA-rats, but not in female BPA-rats, in comparison with vehicle-injected controls. Following mild-stressor the elevation of corticosterone or ACTH levels was higher in male BPA-rats, whereas it was lower in female BPA-rats than vehicle-injected controls. In comparison with vehicle-injected controls, the level of glucocorticoid receptor (GR) mRNA in hippocampus or hypothalamic paraventricular nucleus was increased in female BPA-rats, while decreased in male BPA-rats. In addition, the levels of hippocampal mineralocorticoid receptor (MR) mRNA, neuronal nitric oxide synthase (nNOS) and phospho-cAMP response element binding protein (p-CREB) were increased in female BPA-rats, but were decreased in male BPA-rats. Furthermore, the testosterone level was reduced in male BPA-rats. The results indicate that the perinatal exposure to BPA through altering the GR and MR expression disrupts the GR-mediated feedback of hypothalamic-pituitary-adrenal (HPA) axis and MR-induced nNOS-CREB signaling, which alters sex difference in affective behaviors.
Article
Mutations in Prop1 account for up to half of the cases of combined pituitary hormone deficiency that result from known causes. Despite this, few signaling molecules and pathways that influence Prop1 expression have been identified. Notch signaling has been linked to Prop1 expression, but the developmental periods during which Notch signaling influences Prop1 and overall pituitary development remain unclear. To test the requirement for Notch signaling in establishing the normal pituitary hormone milieu, we generated mice with early embryonic conditional loss of Notch2 (cKO) and examined the consequences of chemical Notch inhibition during early postnatal pituitary maturation. We show that loss of Notch2 has little influence on early embryonic pituitary proliferation, but is crucial for postnatal progenitor maintenance and proliferation. Additionally, we show that Notch signaling is necessary embryonically and postnatally for Prop1 expression and robust Pit1 lineage hormone cell expansion, as well as repression of the corticotrope lineage. Taken together, our studies identify temporal and cell-type specific roles for Notch signaling and highlight the importance of this pathway throughout pituitary development.
Article
Bisphenol A (BPA) is a known reproductive toxicant in rodents. However, the effects of in utero BPA exposure on early ovarian development and the consequences of such exposure on female reproduction in later reproductive life are unclear. Thus, we determined the effects of in utero BPA exposure during a critical developmental window on germ cell nest breakdown, a critical process for establishment of the finite primordial follicle pool, and on female reproduction. Pregnant FVB mice (F0) were orally dosed daily with tocopherol-striped corn oil (vehicle), diethylstilbestrol (DES; 0.05 μg/kg, positive control), or BPA (0.5, 20, and 50μg/kg) from gestational day 11 until birth. Ovarian morphology and gene expression profiles then were examined in F1 female offspring on postnatal day (PND) 4 and estrous cyclicity was examined daily after weaning for 30 days. F1 females were also subjected to breeding studies with untreated males at three to nine months. The results indicate that BPA inhibits germ cell nest breakdown via altering expression of selected apoptotic factors. BPA also significantly advances the age of first estrus, shortens the time that the females remain in estrus, and increases the time the females remain in metestrus and diestrus compared to controls. Further, F1 females exposed to low doses of BPA exhibit various fertility problems and have a significantly higher percentage of dead pups compared to controls. These results indicate that in utero exposure to low doses of BPA during a critical ovarian developmental window interferes with early ovarian development and reduces fertility with age.