Abstract

Gestational exposure to the estrogenic endocrine disruptor methoxychlor (MXC) disrupts the female reproductive system at the molecular, physiological, and behavioral levels in adulthood. The current study addressed whether perinatal exposure to endocrine disruptors reprograms expression of a suite of genes expressed in the hypothalamus that control reproductive function and related these molecular changes to premature reproductive aging. Fischer rats were exposed daily for 12 consecutive days to vehicle (dimethylsulfoxide), estradiol benzoate (EB) (1 mg/kg), and MXC (low dose, 20 μg/kg or high dose, 100 mg/kg), beginning on embryonic d 19 through postnatal d 7. The perinatally exposed females were aged to 16–17 months and monitored for reproductive senescence. After euthanasia, hypothalamic regions [preoptic area (POA) and medial basal hypothalamus] were dissected for real-time PCR of gene expression or pyrosequencing to assess DNA methylation of the Esr1 gene. Using a 48-gene PCR platform, two genes (Kiss1 and Esr1) were significantly different in the POA of endocrine-disrupting chemical-exposed rats compared with vehicle-exposed rats after Bonferroni correction. Fifteen POA genes were up-regulated by at least 50% in EB or high-dose MXC compared with vehicle. To understand the epigenetic basis of the increased Esr1 gene expression, we performed bisulfite conversion and pyrosequencing of the Esr1 promoter. EB-treated rats had significantly higher percentage of methylation at three CpG sites in the Esr1 promoter compared with control rats. Together with these molecular effects, perinatal MXC and EB altered estrous cyclicity and advanced reproductive senescence. Thus, early life exposure to endocrine disruptors has lifelong effects on neuroendocrine gene expression and DNA methylation, together with causing the advancement of reproductive senescence.

The control of reproductive aging in female mammals involves a complex interplay of the hypothalamus, pituitary, and ovary. The hypothalamus plays an important role in this life transition, with hypothalamic GnRH neurons changing the synthesis and release of the decapeptide with aging in a species-specific manner (1, 2). Throughout the life cycle, regulatory inputs to GnRH neurons from neuropeptides (e.g. kisspeptin), neurotransmitters [e.g. glutamate and γ-aminobutyric acid (GABA)], and neurotrophic factors (e.g. TGF and IGF) control GnRH gene expression and release, and the balance of these inputs determine the final output of GnRH release from nerve terminals into the portal capillary system that vascularizes the anterior pituitary gland (36). In addition, feedback from peripheral hormones, such as sex steroids (7, 8), regulates hypothalamic output via actions of estradiol, progesterone, and androgens on their respective steroid hormone receptors that are abundant in the hypothalamus. This complex network of neurons, glia, and steroid regulation changes substantially during reproductive aging (912). Nevertheless, relatively little is known about how the hypothalamus changes with age, and its cause-and-effect role on reproductive senescence.

The timing and progression of reproductive senescence is determined by a combination of genetic and environmental factors. To date, most research has focused on genetic predispositions (13). Nevertheless, the environment is likely to play a role, and there is speculation that endocrine-disrupting chemicals (EDC) may hasten reproductive aging, with the potential outcome of shortening an individual's reproductive lifespan. EDC are compounds in the environment that act upon the body's hormonal systems and include industrial contaminants, plastics/plasticizers, pesticides, and other compounds (14). Recent studies show that exposures to EDC during key developmental periods, especially prenatal/early postnatal life, can cause molecular/cellular changes that affect the function of the affected tissues later in life, a concept referred to as the fetal/developmental basis of adult disease (15). The mechanisms for these effects are diverse and probably involve epigenetic molecular changes, including DNA methylation and histone modifications, the consequences of which are manifested later in life. Intriguingly, the latency between exposure and adult dysfunction extends into the realm of reproductive aging. Animal studies show that reproductive aging is accelerated by EDC [methoxychlor (MXC) (16), bisphenol A (17), and dioxins (18)], and recent epidemiological evidence links developmental EDC exposures to accelerated menopause [diethylstilbestrol (19) and perfluorocarbons (20)].

The current study sought to draw mechanistic connections between early life developmental exposure to an estrogenic endocrine disruptor, MXC, its actions on the hypothalamus, and the consequences on reproductive senescence. MXC is an organochlorine pesticide with estrogenic and antiestrogenic properties (16, 21, 22). Some studies have investigated effects of MXC on the female hypothalamus (23), but few have taken a mechanistic approach to understanding how MXC may affect hypothalamic gene expression (24, 25), and none has linked early life exposures to reproductive aging. Amid concerns that infertility is on the rise (26, 27), understanding the links between EDC exposures and reproductive aging are critical. Furthermore, such work extends observations on the fetal basis of adult disease beyond the young adult and into the realm of aging. Finally, this study focused on the hypothalamic mechanisms by which EDC may reprogram the neuroendocrine circuit regulating reproduction. Therefore, as a whole, this project's goal was to determine early life programming of the entire life cycle's reproductive capacity and the underlying molecular mechanisms.

Results

Loss of estrous cyclicity is accelerated in perinatal endocrine-disrupted rats

Rats were monitored by daily vaginal smears for a minimum of 12 consecutive days per month, or longer (up to 20 d) if more time was required to establish cyclicity status, during the 5-month period leading up to euthanasia. Table 1 shows that most dimethylsulfoxide (DMSO)-treated rats were still exhibiting regular cycles at 16–17 months of age. The low-dose MXC group had some diminution in regular cycles with aging, but this was only significantly different from the vehicle group at 14 months. By contrast, the high-dose MXC and the estradiol benzoate (EB)-treated groups had few (if any) rats still exhibiting cycles by 13 months of age. Fisher's exact test revealed that these latter groups were significantly different from the control group from 13 to 17 months of age (P < 0.001–0.0001). Thus, perinatal EDC treatment was associated with premature reproductive failure.

Table 1.

Estrous cycle classification for aging rats treated between E19 and P7

13 Months14 Months15 Months16 Months17 Months
DMSO83% Reg100% Reg83% Reg80% Reg100% Reg
n = 6n = 6n = 6n = 5n = 5
20 μg of MXC100% Reg78% Reg*78% Reg66% Reg66% Reg
n = 9n = 9n = 9n = 9n = 9
100 mg of MXC0% Reg**22% Reg**11% Reg**11% Reg**0% Reg**
n = 9n = 9n = 9n = 9n = 9
1 mg of EB0% Reg**0% Reg**0% Reg**0% Reg**0% Reg**
n = 5n = 5n = 5n = 5n = 5
13 Months14 Months15 Months16 Months17 Months
DMSO83% Reg100% Reg83% Reg80% Reg100% Reg
n = 6n = 6n = 6n = 5n = 5
20 μg of MXC100% Reg78% Reg*78% Reg66% Reg66% Reg
n = 9n = 9n = 9n = 9n = 9
100 mg of MXC0% Reg**22% Reg**11% Reg**11% Reg**0% Reg**
n = 9n = 9n = 9n = 9n = 9
1 mg of EB0% Reg**0% Reg**0% Reg**0% Reg**0% Reg**
n = 5n = 5n = 5n = 5n = 5

Fisher's exact test was conducted with significance at *, P < 0.001; **, P < 0.0001 vs. control (DMSO) at the same age. Reg, Regular estrous cycles of 4–5 d; n, sample size at each age.

Table 1.

Estrous cycle classification for aging rats treated between E19 and P7

13 Months14 Months15 Months16 Months17 Months
DMSO83% Reg100% Reg83% Reg80% Reg100% Reg
n = 6n = 6n = 6n = 5n = 5
20 μg of MXC100% Reg78% Reg*78% Reg66% Reg66% Reg
n = 9n = 9n = 9n = 9n = 9
100 mg of MXC0% Reg**22% Reg**11% Reg**11% Reg**0% Reg**
n = 9n = 9n = 9n = 9n = 9
1 mg of EB0% Reg**0% Reg**0% Reg**0% Reg**0% Reg**
n = 5n = 5n = 5n = 5n = 5
13 Months14 Months15 Months16 Months17 Months
DMSO83% Reg100% Reg83% Reg80% Reg100% Reg
n = 6n = 6n = 6n = 5n = 5
20 μg of MXC100% Reg78% Reg*78% Reg66% Reg66% Reg
n = 9n = 9n = 9n = 9n = 9
100 mg of MXC0% Reg**22% Reg**11% Reg**11% Reg**0% Reg**
n = 9n = 9n = 9n = 9n = 9
1 mg of EB0% Reg**0% Reg**0% Reg**0% Reg**0% Reg**
n = 5n = 5n = 5n = 5n = 5

Fisher's exact test was conducted with significance at *, P < 0.001; **, P < 0.0001 vs. control (DMSO) at the same age. Reg, Regular estrous cycles of 4–5 d; n, sample size at each age.

Serum estradiol and progesterone concentrations in endocrine-disrupted rats

Serum hormone levels were measured in terminal blood samples. Figure 1 shows that serum estradiol concentrations in the aged females were affected by perinatal treatment (P < 0.005, Kruskal-Wallis). Levels of estradiol were significantly lower in the EB group than all the other groups (Mann-Whitney post hoc, P < 0.005). Both the low-dose and high-dose MXC groups also differed from the control (P < 0.05). For progesterone, no differences were determined among the groups (P = 0.375).

Serum concentrations of estradiol (A) and progesterone (B) in aging female rats at 16–17 months of age. Levels of estradiol were significantly lower in the EB group compared with all others. Levels were also lower in the two MXC groups compared with DMSO. Serum progesterone concentrations were unaffected by perinatal exposure to EDC. #, P < 0.05 vs. control; *, P < 0.005 vs. all other groups.
Fig. 1.

Serum concentrations of estradiol (A) and progesterone (B) in aging female rats at 16–17 months of age. Levels of estradiol were significantly lower in the EB group compared with all others. Levels were also lower in the two MXC groups compared with DMSO. Serum progesterone concentrations were unaffected by perinatal exposure to EDC. #, P < 0.05 vs. control; *, P < 0.005 vs. all other groups.

Hypothalamic gene expression is reprogrammed in aging, endocrine-disrupted rats

The low-density PCR array was used to measure expression of 48 genes in the preoptic area (POA) and medial basal hypothalamus (MBH) of perinatally exposed rats euthanized at aging endpoints. Figure 2 shows expression of those genes in the POA that differed in the aging rats by 50% or more between the perinatal vehicle- and EDC-treated groups and based on raw P values of less than 0.05 before Bonferroni correction. Seventeen genes were differentially regulated by perinatal EDC treatments according to these criteria, including steroid hormone receptors/coregulatory factors/binding partners (Esr1, AR, Pgr, Srd5a1, Sts, and Arnt); glutamate/GABA receptor subunits (Gria3, Grin2b, Grik2, and Gabbr1); neurotrophic factors/receptors (Tgfa, Tgfb1, and Igf1r); neuropeptides/receptors (Kiss1, Kiss1r, and Gnrhr); and the transcription factor, Stat5b. Of these 17 genes, 15 showed a similar pattern of expression in the aging rats, with mRNA levels higher in the EB and/or MXC high-dose groups compared with vehicle. Two genes had unique expression patterns. Kiss1 mRNA levels were significantly lower in the EB group compared with all other groups (P < 0.001). Gnrhr mRNA levels were higher in the high-dose MXC group compared with all others (P < 0.05, nonsignificant after Bonferroni correction). After Bonferroni correction, the two POA genes significantly affected by perinatal EDC treatment were: Esr1 (P ≤ 0.001) and Kiss1 (P < 0.001) (Fig. 2).

Low-density PCR analysis identified 48 transcripts in the POA, and data shown for those transcripts with raw P < 0.05 grouped into 5 classes: A) steroid hormone signaling; B) neurotransmitter signaling; C) cytokine signaline; D) neuropeptide signaling; and E) transcription factors. 17 genes met these criteria, and of those, 15 had expression patterns with levels higher in the EB and/or MXC high-dose group compared with DMSO. For this group, Esr1 gene expression met significance at P < 0.001 (DMSO and MXC low dose were both significantly lower than EB and MXC high dose). Two genes had unique expression patterns. Kiss1 gene expression was significant at P < 0.001, with levels significantly lower in the EB group compared with all others. Gnrhr expression had highest levels in the MXC high-dose group. Raw P values are shown for each gene. Gene expression data for the remaining POA genes (P > 0.05) are presented in Supplemental Table 1.
Fig. 2.

Low-density PCR analysis identified 48 transcripts in the POA, and data shown for those transcripts with raw P < 0.05 grouped into 5 classes: A) steroid hormone signaling; B) neurotransmitter signaling; C) cytokine signaline; D) neuropeptide signaling; and E) transcription factors. 17 genes met these criteria, and of those, 15 had expression patterns with levels higher in the EB and/or MXC high-dose group compared with DMSO. For this group, Esr1 gene expression met significance at P < 0.001 (DMSO and MXC low dose were both significantly lower than EB and MXC high dose). Two genes had unique expression patterns. Kiss1 gene expression was significant at P < 0.001, with levels significantly lower in the EB group compared with all others. Gnrhr expression had highest levels in the MXC high-dose group. Raw P values are shown for each gene. Gene expression data for the remaining POA genes (P > 0.05) are presented in Supplemental Table 1.

In the MBH, gene expression of only one transcript (solute carrier 17a6, vesicular glutamate transporter 2) was higher in the MXC high-dose group compared with all others (P < 0.05, nonsignificant after Bonferroni correction). Other MBH genes were not altered by perinatal EDC treatments. Supplemental Tables 1 and 2, published on The Endocrine Society's Journals Online web site at http://mend.endojournals.org, show relative gene expression of those genes that were not affected in either POA (Supplemental Table 1) and all genes in MBH (Supplemental Table 2).

DNA methylation of estrogen receptor (ER)α (Esr1) gene by perinatal EDC

Because the Esr1 gene was significantly up-regulated in the brain of aging females, we performed bisulfite conversion and pyrosequencing of two sites of the Esr1 regulatory regions in exon 1b and intron 1 to determine whether long-term epigenetic programming of the Esr1 gene had occurred. These regions were chosen because DNA methylation of CpG sites in these two gene regions is regulated by maternal behavior or perinatal estrogen when assessed in young adults (2830). Fourteen CpG sites in exon 1b, and 11 sites in intron 1, were analyzed by pyrosequencing for percentage of methylation. The total percentage of methylation of all the CpG sites within the two regions of analysis was unaffected by perinatal EDC treatment (Fig. 3). The sequences and location of CpG sites of the regions of analysis are also shown in Fig. 3. When individual CpG sites were examined for percentage of methylation, one site in exon 1b (+46 from the transcription start site) and two sites in intron 1 (+1733 and +1786 from the transcription start site) were up-regulated in the perinatally EB-treated rats compared with controls (Fig. 4).

Methylation of CpG sites in the Esr1 gene was assayed by bisulfite reaction and pyrosequencing. For exon 1b (A), total methylation across the 14 sites analyzed was not significantly altered by the perinatal EDC exposure. Similarly, total methylation of 11 sites in intron 1 (B) was unaffected by treatment. The regions of analysis are shown in C (exon 1b) and D (intron 1). In C and D, CpG sites are highlighted in gray. The transcription start site in exon 1b is indicated by bold underlined text. Numbers to the left of the sequence (B and C) are nucleotide positions relative to the transcription start site in exon 1b.
Fig. 3.

Methylation of CpG sites in the Esr1 gene was assayed by bisulfite reaction and pyrosequencing. For exon 1b (A), total methylation across the 14 sites analyzed was not significantly altered by the perinatal EDC exposure. Similarly, total methylation of 11 sites in intron 1 (B) was unaffected by treatment. The regions of analysis are shown in C (exon 1b) and D (intron 1). In C and D, CpG sites are highlighted in gray. The transcription start site in exon 1b is indicated by bold underlined text. Numbers to the left of the sequence (B and C) are nucleotide positions relative to the transcription start site in exon 1b.

Methylation of individual CpG sites in exon 1b (A) and intron 1 (B) of Esr1 is shown for the four perinatal treatment groups. In exon 1b, one site located at +46 relative to the transcription start site in exon 1 differed by perinatal treatment, with levels significantly higher in the EB group compared with DMSO. In intron 1, two sites located at +1733 and +1786 were also affected, again with the EB group having significantly higher percentage of methylation compared with DMSO.
Fig. 4.

Methylation of individual CpG sites in exon 1b (A) and intron 1 (B) of Esr1 is shown for the four perinatal treatment groups. In exon 1b, one site located at +46 relative to the transcription start site in exon 1 differed by perinatal treatment, with levels significantly higher in the EB group compared with DMSO. In intron 1, two sites located at +1733 and +1786 were also affected, again with the EB group having significantly higher percentage of methylation compared with DMSO.

Putative transcription factors in exon 1b and intron 1 of Esr1

MatInspector analysis of the exon 1b and intron 1 sequences of the ERα gene revealed numerous putative transcription factor sites on the forward (+) and reverse (−) strands. Table 2 shows the list of transcription factors with more than 80% similarity to the prototypical sequences in the 221-nucleotide sequence assayed for exon 1b. Forty putative transcription factors with homology to the rat prototypical sequences were identified, with similar distribution on the forward (+) and reverse (−) DNA strand. There were seven potential binding sites for six transcription factors (one transcription factor had two sites) in the hypermethylated +46 region.

Table 2.

Transcription factors in the exon 1b sequence with more than 80% homology to the rat prototypical sequences

FamilyDescriptionPosition (from)Position (to)AnchorStrandMatrix similarity
YBXFY-box binding transcription factor−143−131−137(−)0.889
ETSFHuman/murine ETS factors−134−114−124(−)0.855
NF1FNuclear factor 1−130−110−120(+)0.929
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors−128−104−116(−)0.925
MYODMyoblast determining factors−127−111−119(−)0.911
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors−125−101−113(−)0.984
HOXFParalog hox genes 1–8 from the four hox clusters A, B, C, D−123−105−114(−)0.966
YY1FActivator/repressor binding to transcription initiation site−121−101−111(+)0.877
WHNFWinged helix binding sites−109−99−104(−)0.974
AP4RAP4 and related proteins−94−78−86(+)0.851
MYODMyoblast determining factors−94−78−86(−)0.928
NEURNeuroD, Beta2, HLH domain pancreas transcription factor 1−93−81−87(+)0.964
CTCFCTCF and BORIS gene family transcrip-tional regulators−82−56−69(+)0.843
EGRFEGR/NGF induced protein C and related factors−78−62−70(+)0.888
SP1FGC-box factors SP1/GC−77−61−69(+)0.910
MAXFMyc associated zinc fingers−74−62−68(+)0.913
KLFSKrueppel like transcription factors−74−58−66(+)0.824
HESFVertebrate homologs of enhancer of split complex−65−51−58(−)0.870
EBOXE-box binding factors−64−52−58(+)0.912
EBOXE-box binding factors−63−51−57(−)0.928
LTFMLactotransferrin motif−61−53−57(−)0.984
CP2FCP2-erythrocyte factor related to Drosophila Elf1−47−29−38(+)0.881
NOLFNeuron-specific olfactory factor−42−20−31(+)0.888
PRDMPRD1-BF1 and RIZ homologous (PR) domain proteins (PRDM)−42−13−27(−)0.802
NFKBNuclear factor κ B/c-rel−36−22−29(−)0.824
FASTFAST-1 SMAD interacting proteins−23−7−15(−)0.851
FKHDForkhead domain factors−152−7(+)0.926
EGRFEGR/NGF induced protein C and related factors−3146(−)0.808
SP1FGC-box factors SP1/GC193527(−)0.879
CDEFCell cycle regulators: Cell cycle dependent element243630(−)0.949
E2FFE2F-myc activator/cell cycle regulator254133(+)0.864
TF2BRNA polymerase II transcription factor IIB283431(+)1.000
HOMFHomeodomain transcription factors314940(+)0.899
ZFO3C2H2 zinc finger transcription factors 3344640(−)0.916
HEATHeat shock factors355947(−)0.953
HEATHeat shock factors366048(+)0.886
INRECore promotor initiator elements394944(+)0.945
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors396351(−)0.872
HOXFParalog hox genes 1–8 from the four hox clusters A, B, C, D446253(−)0.942
TEAFTEA/ATTS DNA binding domain factors496155(+)0.967
FamilyDescriptionPosition (from)Position (to)AnchorStrandMatrix similarity
YBXFY-box binding transcription factor−143−131−137(−)0.889
ETSFHuman/murine ETS factors−134−114−124(−)0.855
NF1FNuclear factor 1−130−110−120(+)0.929
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors−128−104−116(−)0.925
MYODMyoblast determining factors−127−111−119(−)0.911
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors−125−101−113(−)0.984
HOXFParalog hox genes 1–8 from the four hox clusters A, B, C, D−123−105−114(−)0.966
YY1FActivator/repressor binding to transcription initiation site−121−101−111(+)0.877
WHNFWinged helix binding sites−109−99−104(−)0.974
AP4RAP4 and related proteins−94−78−86(+)0.851
MYODMyoblast determining factors−94−78−86(−)0.928
NEURNeuroD, Beta2, HLH domain pancreas transcription factor 1−93−81−87(+)0.964
CTCFCTCF and BORIS gene family transcrip-tional regulators−82−56−69(+)0.843
EGRFEGR/NGF induced protein C and related factors−78−62−70(+)0.888
SP1FGC-box factors SP1/GC−77−61−69(+)0.910
MAXFMyc associated zinc fingers−74−62−68(+)0.913
KLFSKrueppel like transcription factors−74−58−66(+)0.824
HESFVertebrate homologs of enhancer of split complex−65−51−58(−)0.870
EBOXE-box binding factors−64−52−58(+)0.912
EBOXE-box binding factors−63−51−57(−)0.928
LTFMLactotransferrin motif−61−53−57(−)0.984
CP2FCP2-erythrocyte factor related to Drosophila Elf1−47−29−38(+)0.881
NOLFNeuron-specific olfactory factor−42−20−31(+)0.888
PRDMPRD1-BF1 and RIZ homologous (PR) domain proteins (PRDM)−42−13−27(−)0.802
NFKBNuclear factor κ B/c-rel−36−22−29(−)0.824
FASTFAST-1 SMAD interacting proteins−23−7−15(−)0.851
FKHDForkhead domain factors−152−7(+)0.926
EGRFEGR/NGF induced protein C and related factors−3146(−)0.808
SP1FGC-box factors SP1/GC193527(−)0.879
CDEFCell cycle regulators: Cell cycle dependent element243630(−)0.949
E2FFE2F-myc activator/cell cycle regulator254133(+)0.864
TF2BRNA polymerase II transcription factor IIB283431(+)1.000
HOMFHomeodomain transcription factors314940(+)0.899
ZFO3C2H2 zinc finger transcription factors 3344640(−)0.916
HEATHeat shock factors355947(−)0.953
HEATHeat shock factors366048(+)0.886
INRECore promotor initiator elements394944(+)0.945
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors396351(−)0.872
HOXFParalog hox genes 1–8 from the four hox clusters A, B, C, D446253(−)0.942
TEAFTEA/ATTS DNA binding domain factors496155(+)0.967

Positions are shown relative to the transcription start site in exon 1b. (+), Forward; (−), reverse.

Table 2.

Transcription factors in the exon 1b sequence with more than 80% homology to the rat prototypical sequences

FamilyDescriptionPosition (from)Position (to)AnchorStrandMatrix similarity
YBXFY-box binding transcription factor−143−131−137(−)0.889
ETSFHuman/murine ETS factors−134−114−124(−)0.855
NF1FNuclear factor 1−130−110−120(+)0.929
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors−128−104−116(−)0.925
MYODMyoblast determining factors−127−111−119(−)0.911
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors−125−101−113(−)0.984
HOXFParalog hox genes 1–8 from the four hox clusters A, B, C, D−123−105−114(−)0.966
YY1FActivator/repressor binding to transcription initiation site−121−101−111(+)0.877
WHNFWinged helix binding sites−109−99−104(−)0.974
AP4RAP4 and related proteins−94−78−86(+)0.851
MYODMyoblast determining factors−94−78−86(−)0.928
NEURNeuroD, Beta2, HLH domain pancreas transcription factor 1−93−81−87(+)0.964
CTCFCTCF and BORIS gene family transcrip-tional regulators−82−56−69(+)0.843
EGRFEGR/NGF induced protein C and related factors−78−62−70(+)0.888
SP1FGC-box factors SP1/GC−77−61−69(+)0.910
MAXFMyc associated zinc fingers−74−62−68(+)0.913
KLFSKrueppel like transcription factors−74−58−66(+)0.824
HESFVertebrate homologs of enhancer of split complex−65−51−58(−)0.870
EBOXE-box binding factors−64−52−58(+)0.912
EBOXE-box binding factors−63−51−57(−)0.928
LTFMLactotransferrin motif−61−53−57(−)0.984
CP2FCP2-erythrocyte factor related to Drosophila Elf1−47−29−38(+)0.881
NOLFNeuron-specific olfactory factor−42−20−31(+)0.888
PRDMPRD1-BF1 and RIZ homologous (PR) domain proteins (PRDM)−42−13−27(−)0.802
NFKBNuclear factor κ B/c-rel−36−22−29(−)0.824
FASTFAST-1 SMAD interacting proteins−23−7−15(−)0.851
FKHDForkhead domain factors−152−7(+)0.926
EGRFEGR/NGF induced protein C and related factors−3146(−)0.808
SP1FGC-box factors SP1/GC193527(−)0.879
CDEFCell cycle regulators: Cell cycle dependent element243630(−)0.949
E2FFE2F-myc activator/cell cycle regulator254133(+)0.864
TF2BRNA polymerase II transcription factor IIB283431(+)1.000
HOMFHomeodomain transcription factors314940(+)0.899
ZFO3C2H2 zinc finger transcription factors 3344640(−)0.916
HEATHeat shock factors355947(−)0.953
HEATHeat shock factors366048(+)0.886
INRECore promotor initiator elements394944(+)0.945
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors396351(−)0.872
HOXFParalog hox genes 1–8 from the four hox clusters A, B, C, D446253(−)0.942
TEAFTEA/ATTS DNA binding domain factors496155(+)0.967
FamilyDescriptionPosition (from)Position (to)AnchorStrandMatrix similarity
YBXFY-box binding transcription factor−143−131−137(−)0.889
ETSFHuman/murine ETS factors−134−114−124(−)0.855
NF1FNuclear factor 1−130−110−120(+)0.929
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors−128−104−116(−)0.925
MYODMyoblast determining factors−127−111−119(−)0.911
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors−125−101−113(−)0.984
HOXFParalog hox genes 1–8 from the four hox clusters A, B, C, D−123−105−114(−)0.966
YY1FActivator/repressor binding to transcription initiation site−121−101−111(+)0.877
WHNFWinged helix binding sites−109−99−104(−)0.974
AP4RAP4 and related proteins−94−78−86(+)0.851
MYODMyoblast determining factors−94−78−86(−)0.928
NEURNeuroD, Beta2, HLH domain pancreas transcription factor 1−93−81−87(+)0.964
CTCFCTCF and BORIS gene family transcrip-tional regulators−82−56−69(+)0.843
EGRFEGR/NGF induced protein C and related factors−78−62−70(+)0.888
SP1FGC-box factors SP1/GC−77−61−69(+)0.910
MAXFMyc associated zinc fingers−74−62−68(+)0.913
KLFSKrueppel like transcription factors−74−58−66(+)0.824
HESFVertebrate homologs of enhancer of split complex−65−51−58(−)0.870
EBOXE-box binding factors−64−52−58(+)0.912
EBOXE-box binding factors−63−51−57(−)0.928
LTFMLactotransferrin motif−61−53−57(−)0.984
CP2FCP2-erythrocyte factor related to Drosophila Elf1−47−29−38(+)0.881
NOLFNeuron-specific olfactory factor−42−20−31(+)0.888
PRDMPRD1-BF1 and RIZ homologous (PR) domain proteins (PRDM)−42−13−27(−)0.802
NFKBNuclear factor κ B/c-rel−36−22−29(−)0.824
FASTFAST-1 SMAD interacting proteins−23−7−15(−)0.851
FKHDForkhead domain factors−152−7(+)0.926
EGRFEGR/NGF induced protein C and related factors−3146(−)0.808
SP1FGC-box factors SP1/GC193527(−)0.879
CDEFCell cycle regulators: Cell cycle dependent element243630(−)0.949
E2FFE2F-myc activator/cell cycle regulator254133(+)0.864
TF2BRNA polymerase II transcription factor IIB283431(+)1.000
HOMFHomeodomain transcription factors314940(+)0.899
ZFO3C2H2 zinc finger transcription factors 3344640(−)0.916
HEATHeat shock factors355947(−)0.953
HEATHeat shock factors366048(+)0.886
INRECore promotor initiator elements394944(+)0.945
SORYSOX/SRY-sex/testis-determining factors and related HMG box factors396351(−)0.872
HOXFParalog hox genes 1–8 from the four hox clusters A, B, C, D446253(−)0.942
TEAFTEA/ATTS DNA binding domain factors496155(+)0.967

Positions are shown relative to the transcription start site in exon 1b. (+), Forward; (−), reverse.

In the 174 nucleotide sequence of intron 1 (Table 3), 33 transcription factors were found that met the 80% homology cut-off. A cluster of 13 overlapping transcription factors distributed across the (+) and (−) DNA strand was found in the 20-nucleotide region from 1827 to 1846 nucleotides downstream from the transcription start site in exon 1b, a region characterized by four adjacent repeating CpG sites (see Fig. 3). Within that region, five putative ZF5 POZ domain zinc finger, four E2F-myc activator/cell cycle regulator, two nuclear respiratory factor 1 (NRF1), one cell cycle-dependent element, and one signal transducer and activator of transcription (STAT) sites were identified. The intron 1 sequence also contained a total of five STAT sites in the region of analysis.

Table 3.

Transcription factors in the intron 1 sequence with more than 80% homology to the rat prototypical sequences

FamilyDescriptionPosition (from)Position (to)AnchorStrandMatrix similarity
E2FFE2F-myc activator/cell cycle regulator174617621754(−)0.815
PLAGPleomorphic adenoma gene174717691758(−)1.000
KLFSKrueppel like transcription factors175717731765(−)0.928
INSMInsulinoma associated factors175917711765(−)0.917
STATSignal transducer and activator of transcription178418021793(−)0.952
HNF1Hepatic nuclear factor 1179218081800(+)0.852
NFATNuclear factor of activated T-cells179218101801(+)0.872
MYT1MYT1 C2HC zinc finger protein179518071801(+)0.967
MYT1MYT1 C2HC zinc finger protein179818101804(−)0.817
STATSignal transducer and activator of transcription180018181809(−)0.910
STATSignal transducer and activator of transcription180218201811(+)0.961
EVI1EVI1-myleoid transforming protein180818241816(+)0.821
GATAGATA binding factors181118231817(+)0.982
OVOLOVO homolog-like transcription factors181218261819(−)0.831
ZF5FZF5 POZ domain zinc finger182718411834(+)0.966
E2FFE2F-myc activator/cell cycle regulator182718431835(−)0.897
NRF1Nuclear respiratory factor 1182718431835(−)0.830
ZF5FZF5 POZ domain zinc finger182818451835(−)0.952
E2FFE2F-myc activator/cell cycle regulator182818451836(+)0.894
ZF5FZF5 POZ domain zinc finger182918441836(+)1.000
NRF1Nuclear respiratory factor 1182918451837(−)0.802
E2FFE2F-myc activator/cell cycle regulator182918451837(−)0.946
ZF5FZF5 POZ domain zinc finger183018441837(−)0.919
E2FFE2F-myc activator/cell cycle regulator183018461838(+)0.908
ZF5FZF5 POZ domain zinc finger183118451838(+)0.967
CDEFCell cycle dependent element183418461840(+)0.906
STATSignal transducer and activator of Transcription184418621853(−)0.934
VTBPVertebrate TATA binding protein factor185118671859(+)0.850
NKXHNKX homeodomain factors185118691860(+)0.964
RUSHSWI/SNF related nucleophosphoproteins with a RING finger DNA binding motif185418641859(−)0.993
STATSignal transducer and activator of transcription185418721863(−)0.980
LTFMLactotransferrin motif185518631859(−)0.910
KLFSKrueppel like transcription factors186918851877(−)0.925
FamilyDescriptionPosition (from)Position (to)AnchorStrandMatrix similarity
E2FFE2F-myc activator/cell cycle regulator174617621754(−)0.815
PLAGPleomorphic adenoma gene174717691758(−)1.000
KLFSKrueppel like transcription factors175717731765(−)0.928
INSMInsulinoma associated factors175917711765(−)0.917
STATSignal transducer and activator of transcription178418021793(−)0.952
HNF1Hepatic nuclear factor 1179218081800(+)0.852
NFATNuclear factor of activated T-cells179218101801(+)0.872
MYT1MYT1 C2HC zinc finger protein179518071801(+)0.967
MYT1MYT1 C2HC zinc finger protein179818101804(−)0.817
STATSignal transducer and activator of transcription180018181809(−)0.910
STATSignal transducer and activator of transcription180218201811(+)0.961
EVI1EVI1-myleoid transforming protein180818241816(+)0.821
GATAGATA binding factors181118231817(+)0.982
OVOLOVO homolog-like transcription factors181218261819(−)0.831
ZF5FZF5 POZ domain zinc finger182718411834(+)0.966
E2FFE2F-myc activator/cell cycle regulator182718431835(−)0.897
NRF1Nuclear respiratory factor 1182718431835(−)0.830
ZF5FZF5 POZ domain zinc finger182818451835(−)0.952
E2FFE2F-myc activator/cell cycle regulator182818451836(+)0.894
ZF5FZF5 POZ domain zinc finger182918441836(+)1.000
NRF1Nuclear respiratory factor 1182918451837(−)0.802
E2FFE2F-myc activator/cell cycle regulator182918451837(−)0.946
ZF5FZF5 POZ domain zinc finger183018441837(−)0.919
E2FFE2F-myc activator/cell cycle regulator183018461838(+)0.908
ZF5FZF5 POZ domain zinc finger183118451838(+)0.967
CDEFCell cycle dependent element183418461840(+)0.906
STATSignal transducer and activator of Transcription184418621853(−)0.934
VTBPVertebrate TATA binding protein factor185118671859(+)0.850
NKXHNKX homeodomain factors185118691860(+)0.964
RUSHSWI/SNF related nucleophosphoproteins with a RING finger DNA binding motif185418641859(−)0.993
STATSignal transducer and activator of transcription185418721863(−)0.980
LTFMLactotransferrin motif185518631859(−)0.910
KLFSKrueppel like transcription factors186918851877(−)0.925

Positions are shown relative to the transcription start site in exon 1b. (+), Forward; (−), reverse.

Table 3.

Transcription factors in the intron 1 sequence with more than 80% homology to the rat prototypical sequences

FamilyDescriptionPosition (from)Position (to)AnchorStrandMatrix similarity
E2FFE2F-myc activator/cell cycle regulator174617621754(−)0.815
PLAGPleomorphic adenoma gene174717691758(−)1.000
KLFSKrueppel like transcription factors175717731765(−)0.928
INSMInsulinoma associated factors175917711765(−)0.917
STATSignal transducer and activator of transcription178418021793(−)0.952
HNF1Hepatic nuclear factor 1179218081800(+)0.852
NFATNuclear factor of activated T-cells179218101801(+)0.872
MYT1MYT1 C2HC zinc finger protein179518071801(+)0.967
MYT1MYT1 C2HC zinc finger protein179818101804(−)0.817
STATSignal transducer and activator of transcription180018181809(−)0.910
STATSignal transducer and activator of transcription180218201811(+)0.961
EVI1EVI1-myleoid transforming protein180818241816(+)0.821
GATAGATA binding factors181118231817(+)0.982
OVOLOVO homolog-like transcription factors181218261819(−)0.831
ZF5FZF5 POZ domain zinc finger182718411834(+)0.966
E2FFE2F-myc activator/cell cycle regulator182718431835(−)0.897
NRF1Nuclear respiratory factor 1182718431835(−)0.830
ZF5FZF5 POZ domain zinc finger182818451835(−)0.952
E2FFE2F-myc activator/cell cycle regulator182818451836(+)0.894
ZF5FZF5 POZ domain zinc finger182918441836(+)1.000
NRF1Nuclear respiratory factor 1182918451837(−)0.802
E2FFE2F-myc activator/cell cycle regulator182918451837(−)0.946
ZF5FZF5 POZ domain zinc finger183018441837(−)0.919
E2FFE2F-myc activator/cell cycle regulator183018461838(+)0.908
ZF5FZF5 POZ domain zinc finger183118451838(+)0.967
CDEFCell cycle dependent element183418461840(+)0.906
STATSignal transducer and activator of Transcription184418621853(−)0.934
VTBPVertebrate TATA binding protein factor185118671859(+)0.850
NKXHNKX homeodomain factors185118691860(+)0.964
RUSHSWI/SNF related nucleophosphoproteins with a RING finger DNA binding motif185418641859(−)0.993
STATSignal transducer and activator of transcription185418721863(−)0.980
LTFMLactotransferrin motif185518631859(−)0.910
KLFSKrueppel like transcription factors186918851877(−)0.925
FamilyDescriptionPosition (from)Position (to)AnchorStrandMatrix similarity
E2FFE2F-myc activator/cell cycle regulator174617621754(−)0.815
PLAGPleomorphic adenoma gene174717691758(−)1.000
KLFSKrueppel like transcription factors175717731765(−)0.928
INSMInsulinoma associated factors175917711765(−)0.917
STATSignal transducer and activator of transcription178418021793(−)0.952
HNF1Hepatic nuclear factor 1179218081800(+)0.852
NFATNuclear factor of activated T-cells179218101801(+)0.872
MYT1MYT1 C2HC zinc finger protein179518071801(+)0.967
MYT1MYT1 C2HC zinc finger protein179818101804(−)0.817
STATSignal transducer and activator of transcription180018181809(−)0.910
STATSignal transducer and activator of transcription180218201811(+)0.961
EVI1EVI1-myleoid transforming protein180818241816(+)0.821
GATAGATA binding factors181118231817(+)0.982
OVOLOVO homolog-like transcription factors181218261819(−)0.831
ZF5FZF5 POZ domain zinc finger182718411834(+)0.966
E2FFE2F-myc activator/cell cycle regulator182718431835(−)0.897
NRF1Nuclear respiratory factor 1182718431835(−)0.830
ZF5FZF5 POZ domain zinc finger182818451835(−)0.952
E2FFE2F-myc activator/cell cycle regulator182818451836(+)0.894
ZF5FZF5 POZ domain zinc finger182918441836(+)1.000
NRF1Nuclear respiratory factor 1182918451837(−)0.802
E2FFE2F-myc activator/cell cycle regulator182918451837(−)0.946
ZF5FZF5 POZ domain zinc finger183018441837(−)0.919
E2FFE2F-myc activator/cell cycle regulator183018461838(+)0.908
ZF5FZF5 POZ domain zinc finger183118451838(+)0.967
CDEFCell cycle dependent element183418461840(+)0.906
STATSignal transducer and activator of Transcription184418621853(−)0.934
VTBPVertebrate TATA binding protein factor185118671859(+)0.850
NKXHNKX homeodomain factors185118691860(+)0.964
RUSHSWI/SNF related nucleophosphoproteins with a RING finger DNA binding motif185418641859(−)0.993
STATSignal transducer and activator of transcription185418721863(−)0.980
LTFMLactotransferrin motif185518631859(−)0.910
KLFSKrueppel like transcription factors186918851877(−)0.925

Positions are shown relative to the transcription start site in exon 1b. (+), Forward; (−), reverse.

Body weight is affected by perinatal EDC

Rats were weighed on the day of euthanasia. As shown in Fig. 5, the EB group had a significantly higher body weight than rats in all of the other treatment groups (P < 0.005). Post hoc analysis (Fisher's least significant difference) showed that the EB group was significantly heavier than all other groups (P < 0.01 vs. all).

Body weight (g) is shown for rats on the day of euthanasia at 16–17 months of age. The EB group had significantly heavier body weights than all other groups. *, P < 0.01 vs. all other treatments.
Fig. 5.

Body weight (g) is shown for rats on the day of euthanasia at 16–17 months of age. The EB group had significantly heavier body weights than all other groups. *, P < 0.01 vs. all other treatments.

Discussion

The current data show that early life exposures to environmental endocrine disruptors hasten premature reproductive aging, thereby diminishing an individual's lifetime reproductive capacity. Whether this is the case in women remains to be proven, although epidemiological data suggest that certain EDCs are associated with earlier age at menopause. Women who had been exposed to high levels of dioxin due to a chemical plant explosion in Seveso, Italy, showed increased risk for early menopause (31). Body burden of DDE (dichlorodiphenyldichloroethylene), a metabolite of the organochlorine pesticide DDT (dichlorodiphenyltrichloroethane), was associated with earlier age at menopause in a population of women studied in North Carolina (32). A cross-sectional study conducted on Hispanic women relating their body burdens of organochlorine pesticides to age at menopause showed that menopause occurred at earlier ages in women exposed to DDT, DDE, β-hexachlorocyclohexane, and trans-nonachlor (33). These results are consistent with the current study's results.

By contrast, other human studies show no relationship between serum EDC and age at menopause (polychlorinated biphenyl, Refs. 32, 34), and one study indicated that on average, menopause occurred 3 months later in women handling pesticides than in controls (35). To our knowledge, links between MXC and menopause have not been studied in humans.

There are plausible explanations for these different outcomes across the human data, not the least of which are differences in the nature of the EDC studied. Humans (and wildlife) are exposed to complex mixtures of environmental contaminants, and exposures occur throughout the life cycles. Although epidemiological studies are valuable, the ability to draw causal connections between exposure and outcome is not possible. Notably, authors on both sides of this literature make the point that there is not a linear dose-response relationship between body burden and biological outcome. Thus, differences among studies may also result from making inferences across different exposure levels that may not be comparable from one population to another. Last, it is becoming more widely accepted that exposures to EDCs during critical developmental windows may have latent effects (14). However, to draw this kind of connection in humans, it is necessary to relate secular trends in the timing of menopause to exposures received 40–50 yr ago. Thus, animal studies are necessary to understand causal relationships and to discern the underlying mechanisms.

Developmental EDC exposures hasten reproductive senescence

Our current results and others show that the reproductive life cycle, which includes puberty, adult reproductive function, and aging, is significantly altered by perinatal EDC exposures. Estrogenic EDCs, such as MXC, cause advancements in the timing of puberty in females (16; reviewed in Ref. 36). This earlier timing of puberty, however, is accompanied by a diminution of fertility in perinatally treated animals when those rats were examined at d 50–60 (16). Together with results showing earlier reproductive senescence, these results as a whole indicate that the reproductive life cycle is substantially shorter in EDC-exposed animals. This finding is consistent with results for bisphenol A (17), dioxins (18), and others (reviewed in Ref. 35) but not heretofore studied from the hypothalamic molecular approach.

The control of natural reproductive aging, and disruption of these processes by EDC resulting in premature reproductive senescence, involves coordinated physiological processes regulated by the hypothalamic-pituitary-gonadal (HPG) axis. At least some of the reproductive aging process caused by MXC is due to actions on the ovary. Perinatal exposure disrupted folliculogenesis, leading to an increased number of preantral and early antral follicles that failed to achieve maturation and ovulation, and fewer/no corpora lutea were detectable (16). This was accompanied by cellular/molecular changes to the ovary. Other EDCs have been shown to have ovarian follicular effects, including polychlorinated biphenyls (37) and bisphenol A (17, 38). However, these animals still have a follicular pool, making it likely that the hypothalamus is not only a target of perinatal MXC but that its reprogramming hastens reproductive aging.

Developmental reprogramming of the hypothalamus

Hypothalamic function is vulnerable to perinatal EDC actions (reviewed in Ref. 39), and our current data add novel information about the molecular mechanisms by which this occurs. We show lifelong changes in hypothalamic gene expression associated with premature reproductive aging, in a region-specific manner. In the POA of rats, a complex regulatory neural/glial network controlling HPG reproductive function was up-regulated by 50% or more by high-dose MXC or EB treatments, relative to control. The three major steroid hormone receptors (ERα, androgen receptor, and progesterone receptor), glutamate/GABA receptors, growth factors (TGFα and TGFβ), the Kiss1 receptor, and the transcription factor Stat5b showed this pattern of expression. In addition, a key neuropeptide regulating GnRH release, kisspeptin (Kiss1 gene), was down-regulated in the EB group, and the GnRH receptor was up-regulated in the MXC high-dose group. The sum of these changes would be integrated at the level of GnRH output from the hypothalamus to pituitary and subsequently to the gonad. Although GnRH gene expression itself was not altered, this is not surprising in light of evidence that it is probably GnRH release and not gene expression that is most highly regulated, including during reproductive aging (reviewed in Ref. 4).

The two genes significantly affected in the POA by perinatal EDC exposure were Kiss1 (down-regulated in the EB group) and Esr1 (ERα; up-regulated in the EB and MXC high-dose groups). The importance of these two genes on female reproduction, both independently as well as in combination, cannot be understated. Kiss1 neurons in the POA play a crucial role in the capacity for rodents and other species to undergo ovulatory processes (40; reviewed in Ref. 41). This role extends to reproductive aging, which is accompanied by changes in the Kiss1 population as shown in rats, monkeys, and humans, with a concurrent decline in ovulatory capacity (9, 10, 42). Furthermore, the Kiss1 system is vulnerable to EDC exposures (4345). Together with the current work, the hypothalamic Kiss1 system is an important target for future mechanistic studies on how developmental EDC exposures alter hypothalamic function.

The Esr1 gene was significantly up-regulated by EB and high-dose MXC, a finding consistent with previous work showing that estrogenic EDCs can affect hypothalamic Esr1 gene or ERα protein expression (45; reviewed in Ref. 36). The same hypothalamic cells may coexpress Kiss 1 and ERα (46). Recent work has shown that positive feedback regulation of the HPG axis requires coexpression of ERα within Kiss1 neurons (40, 47). These estrogen-sensitive kisspeptinergic neurons are a logical target for further research on endocrine disruptors and are potentially vulnerable to long-term regulation by perinatal exposure.

Lifelong epigenetic changes in Esr1 induced by perinatal EDC

Based on our observation of long-lasting effects of MXC and EB on Esr1 gene expression, together with current literature suggesting that expression of Esr1 is epigenetically regulated in the hypothalamus (28, 30, 48, 49), we found that methylation at three sites (+46 site of exon 1b, +1733 and +1786 sites of intron 1) was increased in the EB group. The MXC rats were not significantly different from control in their DNA methylation pattern, possibly because their reproductive function is still maintained (albeit diminished) with aging. In the EB rats, by contrast, the high dosage given perinatally had profound consequences on reproductive aging, gene expression, and DNA methylation pattern. EB rats also showed higher body weight at death, suggesting that adipogenesis and/or the hypothalamic control of energy balance was dysregulated by the perinatal treatment.

It was surprising to us that both Esr1 mRNA levels and CpG methylation of the Esr1 promoter were increased in the EB group, because the relationship between gene expression and CpG methylation is predicted to be inverse. This has been shown in the brain for Esr1 and for other genes, such as Bdnf (28, 30, 48, 49). Several factors may explain the lack of such a relationship for the EB-treated rats in the current study. First, there are numerous other CpG sites on the Esr1 gene whose methylation pattern was not measured herein but which may play different regulatory roles in Esr1 gene expression. Second, other epigenetic and nonepigenetic regulatory factors (e.g. coregulatory factors and transcription factors) may come into play in the final measure of Esr1 mRNA levels. Third, histone modifications, not measured in this study, can contribute substantially to the epigenetic regulation of gene expression. Fourth, the POA is a highly heterogeneous tissue, with different cell types that may be differentially affected at the gene and protein level by perinatal endocrine disruptors. Fifth, the timing of exposure and the long lag until gene expression measurements may come into play. Indeed, DNA methylation is not fixed throughout the life cycle but may undergo dynamic change postnatally. This has even been shown for Esr1, whose DNA methylation profile undergoes shifts from postnatal d 1 to 20 to 60 in rats (29). Similarly, this concept applies to developmental exposures to EDCs affecting DNA methylation with profiles that undergo dynamic change well into aging in the prostate gland (50). In our rats, this concept could be tested by comparing hypothalamic Esr1 methylation patterns of neonatally endocrine-disrupted rats throughout postnatal life into aging, an area that we are currently investigating.

We scanned the Esr1 sequence for the presence of putative transcription factors to guide future analyses of gene regulatory regions that may be affected by developmental estrogenic EDC exposures. In exon 1b, the hypermethylated CpG at +46 is within the binding sites for several transcription factors, such as SORY, HEAT, and ZFO3, that were shown to play roles as transcriptional repressors (5155), which may relate to the lack of expected inverse correlation between the methylation and gene expression. In intron 1, there was a cluster of transcription factors (ZF5 POZ domain zinc finger, E2F-myc activator/cell cycle regulator, NRF1, cell cycle-dependent element, and STAT) overlying the GC-rich region from +1827 to 1846. The ZF5 transcription factor is best studied for its role as a transcriptional repressor (56). E2F is better associated with transcriptional activation and, more recently, with histone H3K56 acetylation (57). NRF1 is well known for its role in mitochondrial function, but recently has been associated with the mediation of estrogen effects on ERα gene transcription (58), something that may be relevant to the current finding for specific effects of EB on DNA methylation at that site in intron 1. Finally, the intron 1 sequence also contained five STAT sites in our region of analysis, a finding that highlights the potential importance of this transcription factor in Esr1 regulation.

Conclusions

The current findings extend the ever-growing body of research collectively termed the fetal/developmental basis of adult disease to the end of the life spectrum. Our results show that not only do perinatal EDC reprogram hypothalamic gene expression but that they cause long-term changes in epigenetic properties of the Esr1 gene and possibly others not studied herein. These findings have substantial implications for humans. By hastening senescence, EDC may eliminate the possibility of biological children for women who may postpone childbirth for personal or professional reasons. Considering the important roles of estrogens on targets in body and brain, early reproductive senescence may accelerate some disease-related states associated with menopause and affect quality of life in the aging population of women.

Materials and Methods

Animals and husbandry

Fischer (CDF) inbred rats were obtained from Charles River Laboratories (Wilmington, MA) to generate timed-pregnant females. Animals in this study were siblings of those described in Ref. 16 and had identical husbandry in the labs at Rutgers University. Rats were fed a soy-free scientific diet 5V01 rat chow (Lab Diet manufactured by PMI Nutrition International LLC, Brentwood, MO) and tap water ad libitum. They were allowed to develop to 16–17 months of age. During this time, they were subjected to periodic monitoring of estrous cycles by daily vaginal smears (see Ref. 16 for details). The cycles were categorized as regular (4–5 d), prolonged (6+ d), persistent estrus, or persistent diestrus (59). All procedures in the present study were carried out in accordance with the guidelines of the Rutgers University Animal Care and Facilities Committee.

Treatments

Experimental subjects were exposed to one of four treatments (below) for a total of 12 consecutive days. Daily injections began with the pregnant dam (ip) on gestational d 19 through parturition on d 22 (four prenatal days). Then, injections continued with the pups (sc) from postnatal d 0 (birth) to d 7 (eight postnatal days). Treatments were: 1) vehicle control [DMSO sesame oil at 1:2 (three litters, n = 5 individuals)]; 2) EB (1 mg/kg · d) as a positive estrogenic control (three litters, n = 5 individuals); 3) MXC (Sigma, St. Louis, MO), 20 μg/kg · d (low-dose MXC; five litters, n = 9 individuals); and 4) 100 mg/kg · d (high-dose MXC; five litters, n = 9 individuals) in 1 ml of vehicle per kilogram of body weight. The choice of doses of MXC and EB was based on the companion study, in which all animals were treated to cause ovarian disruption (16). The 20 μg/kg · d was selected as an environmentally relevant, low dose of MXC (60, 61). The 100 mg/kg · d MXC is considered a midrange dose, chosen for comparison with published studies (23, 62, 63). The EB dose was selected to be high so as to ensure disruption of both ovary and hypothalamus by an estrogenic compound.

Euthanasia and tissue/blood collection

At 16–17 months of age, female rats were euthanized by decapitation between 1400 and 1600 h. Rats with regular estrous cycles were euthanized on the day of proestrus; on this light cycle, rats are in the preovulatory period of the cycle, when hypothalamic activity is increasing and serum LH and estradiol concentrations are rising. Noncycling rats were euthanized on persistent diestrus or persistent estrus. Trunk blood was collected and serum separated by centrifugation and stored at −80 C. Brains were rapidly removed, and the POA and the MBH were blocked as described previously (64), snap frozen in liquid nitrogen, and stored at −80 C.

RNA extraction and real-time PCR

After shipment to the University of Texas, RNA was extracted and purified, treated with deoxyribonuclease (TURBO DNA kit; Applied Biosystems, Inc., Foster City, CA), and purity and integrity were assessed on the bioanalyzer 2100 (Agilent, Cedar Creek, TX). cDNA conversion was carried out on 2 μg of cytoplasmic RNA (High-capacity cDNA reverse transcription kit; Applied Biosystems, Inc.), aliquoted, and processed for real-time PCR as published (45, 65). A 48-gene TaqMan PCR-based array (Applied Biosystems, Inc.) was designed and used for real-time PCR reactions, which were run on the ABI 7900 real-time PCR machine using parameters described elsewhere (45, 65, 66). Data were analyzed using the normalized Ct (δ-Ct) for each sample before transformation to fold change.

Hormone RIA

Serum 17β-estradiol and progesterone were measured in duplicate 100 μl of serum samples using commercially available RIA kits, according to the manufacturer's instructions (Coat-A-Count RIA kits; Siemens Medical Solutions, Malvern, PA). The assay sensitivity, intra- and interassay coefficients of variance were 8 pg/ml, 5.5 and 6.4% (respectively) for estradiol, and 0.2 ng/ml, 4.6 and 5.9% for progesterone.

DNA methylation analysis of the ERα promoter

During the extraction of RNA described above, the genomic DNA fraction was isolated from the nuclear fraction of the POA and MBH samples used for cytoplasmic RNA isolation. Briefly, nuclear pellets were resuspended in high salt buffer and treated with proteinase K followed by a short (10 min) ribonuclease A treatment. Samples were then extracted with phenol/chloroform and precipitated in 100% isopropanol at −20 C overnight. Pelleted DNA was resuspended in nuclease-free H2O and aliquoted at 20 ng/μl. A 50-μl aliquot for each DNA sample was sent to EpigenDX (Worcester, MA) for bisulfite reaction and pyrosequencing of the ERα regulatory regions in exon 1b and intron 1. Sequences of these regions are shown in Fig. 3.

Transcription factor analysis

Using MatInspector software (Genomatix, Inc., Munich, Germany), the intron 1 and exon 1b sequences used by EpigenDX for DNA methylation analysis were scanned to determine which putative rat transcription factor binding sites were localized in these regions.

Analysis and statistics

Experimental subjects derived from three to five litters per treatment and a total of five to nine rats were used per group. No significant effects were found when birth litter was used as a covariate. Furthermore, intra- and interlitter variability were comparable. Therefore, individual rats were used as the unit of analysis for statistical purposes. ANOVA was used to compare differences among treatment groups. Post hoc analysis was performed (Fisher's projected least significant difference) when a main treatment effect at P < 0.05 was identified. When appropriate, Bonferroni corrections were made for multiple analyses and data reported using Tukey post hoc tests. In a few cases, data did not meet assumptions of homogeneity of variance based on Levene's test and were instead analyzed by the nonparametric Kruskal-Wallis test. For estrous cyclicity, Fisher's exact test was used to compare differences between treated animals with controls.

Acknowledgments

We thank David Crews, Ph.D. (University of Texas at Austin) and David Sweatt, Ph.D. (University of Alabama at Birmingham, AL) for helpful discussions about this article.

This work was supported by National Institutes of Health Grants ES018139 and ES07784 (to A.C.G.), F31 AG034813 (to D.M.W.), and ES013854 (to M.U.) and by the National Institute of Environmental Health Sciences Center Grant ES005022 (to M.U.).

Disclosure Summary: The authors have nothing to disclose.

Abbreviations

     
  • DMSO

    Dimethylsulfoxide

  •  
  • EB

    estradiol benzoate

  •  
  • EDC

    endocrine-disrupting chemical

  •  
  • ER

    estrogen receptor

  •  
  • GABA

    γ-aminobutyric acid

  •  
  • HPG

    hypothalamic-pituitary-gonadal

  •  
  • MBH

    medial basal hypothalamus

  •  
  • MXC

    methoxychlor

  •  
  • NRF1

    nuclear respiratory factor 1

  •  
  • POA

    preoptic area

  •  
  • STAT

    signal transducer and activator of transcription.

References

1.

Gore
AC
,
Windsor-Engnell
BM
,
Terasawa
E

2004
Menopausal increases in pulsatile gonadotropin-releasing hormone release in a nonhuman primate (Macaca mulatta).
Endocrinology
145
:
4653
4659
2.

Rubin
BS
,
Bridges
RS

1989
Alterations in luteinizing hormone-releasing hormone release from the mediobasal hypothalamus of ovariectomized, steroid-primed middle-aged rats as measured by push-pull perfusion.
Neuroendocrinology
49
:
225
232
3.

Maeda
K
,
Ohkura
S
,
Uenoyama
Y
,
Wakabayashi
Y
,
Oka
Y
,
Tsukamura
H
,
Okamura
H

2010
Neurobiological mechanisms underlying GnRH pulse generation by the hypothalamus.
Brain Res
1364
:
103
115
4.

Yin
W
,
Gore
AC

2010
The hypothalamic median eminence and its role in reproductive aging.
Ann NY Acad Sci
1204
:
113
122
5.

Iremonger
KJ
,
Constantin
S
,
Liu
X
,
Herbison
AE

2010
Glutamate regulation of GnRH neuron excitability.
Brain Res
1364
:
35
43
6.

Ojeda
SR
,
Ma
YJ
,
Lee
BJ
,
Prevot
V

2000
Glia-to-neuron signaling and the neuroendocrine control of female puberty.
Recent Prog Horm Res
55
:
197
223
;
discussion 223–224
7.

Wintermantel
TM
,
Campbell
RE
,
Porteous
R
,
Bock
D
,
Gröne
HJ
,
Todman
MG
,
Korach
KS
,
Greiner
E
,
Pérez
CA
,
Schütz
G
,
Herbison
AE

2006
Definition of estrogen receptor pathway critical for estrogen positive feedback to gonadotropin-releasing hormone neurons and fertility.
Neuron
52
:
271
280
8.

Christian
CA
,
Moenter
SM

2010
The neurobiology of preovulatory and estradiol-induced gonadotropin-releasing hormone surges.
Endocr Rev
31
:
544
577
9.

Neal-Perry
G
,
Lebesgue
D
,
Lederman
M
,
Shu
J
,
Zeevalk
GD
,
Etgen
AM

2009
The excitatory peptide kisspeptin restores the luteinizing hormone surge and modulates amino acid neurotransmission in the medial preoptic area of middle-aged rats.
Endocrinology
150
:
3699
3708
10.

Rance
NE

2009
Menopause and the human hypothalamus: evidence for the role of kisspeptin/neurokinin B neurons in the regulation of estrogen negative feedback.
Peptides
30
:
111
122
11.

Chakraborty
TR
,
Gore
AC

2004
Aging-related changes in ovarian hormones, their receptors, and neuroendocrine function.
Exp Biol Med
229
:
977
987
12.

Maffucci
JA
,
Gore
AC

2009
Hypothalamic neural systems controlling the female reproductive life cycle gonadotropin-releasing hormone, glutamate, and GABA.
Int Rev Cell Mol Biol
274
:
69
127
13.

Kok
HS
,
van Asselt
KM
,
van der Schouw
YT
,
Peeters
PH
,
Wijmenga
C

2005
Genetic studies to identify genes underlying menopausal age.
Hum Reprod Update
11
:
483
493
14.

Diamanti-Kandarakis
E
,
Bourguignon
JP
,
Giudice
LC
,
Hauser
R
,
Prins
GS
,
Soto
AM
,
Zoeller
RT
,
Gore
AC

2009
Endocrine-disrupting chemicals: an Endocrine Society scientific statement.
Endocr Rev
30
:
293
342
15.

Barker
DJP

2003
The developmental origins of adult disease.
Eur J Epidemiol
18
:
733
736
16.

Armenti
AE
,
Zama
AM
,
Passantino
L
,
Uzumcu
M

2008
Developmental methoxychlor exposure affects multiple reproductive parameters and ovarian folliculogenesis and gene expression in adult rats.
Toxicol Appl Pharmacol
233
:
286
296
17.

Adewale
HB
,
Jefferson
WN
,
Newbold
RR
,
Patisaul
HB

2009
Neonatal bisphenol-A exposure alters rat reproductive development and ovarian morphology without impairing activation of gonadotropin-releasing hormone neurons.
Biol Reprod
81
:
690
699
18.

Shi
Z
,
Valdez
KE
,
Ting
AY
,
Franczak
A
,
Gum
SL
,
Petroff
BK

2007
Ovarian endocrine disruption underlies premature reproductive senescence following environmentally relevant chronic exposure to the aryl hydrocarbon receptor agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin.
Biol Reprod
76
:
198
202
19.

Hatch
EE
,
Troisi
R
,
Wise
LA
,
Hyer
M
,
Palmer
JR
,
Titus-Ernstoff
L
,
Strohsnitter
W
,
Kaufman
R
,
Adam
E
,
Noller
KL
,
Herbst
AL
,
Robboy
S
,
Hartge
P
,
Hoover
RN

2006
Age at natural menopause in women exposed to diethylstilbestrol in utero.
Am J Epidemiol
164
:
682
688
20.

Knox
SS
,
Jackson
T
,
Javins
B
,
Frisbee
SJ
,
Shankar
A
,
Ducatman
AM

2011
Implications of early menopause in women exposed to perfluorocarbons.
J Clin Endocrinol Metab
96
:
1747
1753
21.

Cummings
AM

1997
Methoxychlor as a model for environmental estrogens.
Crit Rev Toxicol
27
:
367
379
22.

Hall
DL
,
Payne
LA
,
Putnam
JM
,
Huet-Hudson
YM

1997
Effect of methoxychlor on implantation and embryo development in the mouse.
Reprod Toxicol
11
:
703
708
23.

Gray
LEJ
,
Ostby
J
,
Ferrell
J
,
Rehnberg
G
,
Linder
R
,
Cooper
R
,
Goldman
J
,
Slott
V
,
Laskey
J

1989
A dose-response analysis of methoxychlor-induced alterations of reproductive development and function in the rat.
Fund Appl Toxicol
12
:
92
108
24.

Takagi
H
,
Shibutani
M
,
Lee
KY
,
Masutomi
N
,
Fujita
H
,
Inoue
K
,
Mitsumori
K
,
Hirose
M

2005
Impact of maternal dietary exposure to endocrine-acting chemicals on progesterone receptor expression in microdissected hypothalamic medial preoptic areas of rat offspring.
Toxicol Appl Pharmacol
208
:
127
136
25.

Mahoney
MM
,
Padmanabhan
V

2010
Developmental programming: impact of fetal exposure to endocrine-disrupting chemicals on gonadotropin-releasing hormone and estrogen receptor mRNA in sheep hypothalamus.
Toxicol Appl Pharmacol
247
:
98
104
26.

Zama
AM
,
Uzumcu
M

2010
Epigenetic effects of endocrine-disrupting chemicals on female reproduction: an ovarian perspective.
Front Neuroendocrinol
31
:
420
439
27.

Guzick
DS
,
Swan
S

2006
The decline of infertility: apparent or real?
Fertil Steril
86
:
524
526
;
discussion 534
28.

Champagne
FA
,
Weaver
IC
,
Diorio
J
,
Dymov
S
,
Szyf
M
,
Meaney
MJ

2006
Maternal care associated with methylation of the estrogen receptor-α1b promoter and estrogen receptor-α expression in the medial preoptic area of female offspring.
Endocrinology
147
:
2909
2915
29.

Schwarz
JM
,
Nugent
BM
,
McCarthy
MM

2010
Developmental and hormone-induced epigenetic changes to estrogen and progesterone receptor genes in brain are dynamic across the life span.
Endocrinology
151
:
4871
4881
30.

Kurian
JR
,
Olesen
KM
,
Auger
AP

2010
Sex differences in epigenetic regulation of the estrogen receptor-α promoter within the developing preoptic area.
Endocrinology
151
:
2297
2305
31.

Eskenazi
B
,
Warner
M
,
Marks
AR
,
Samuels
S
,
Gerthoux
PM
,
Vercellini
P
,
Olive
DL
,
Needham
L
,
Patterson
D
,
Mocarelli
P

2005
Serum dioxin concentrations and age at menopause.
Environ Health Perspect
113
:
858
862
32.

Cooper
GS
,
Savitz
DA
,
Millikan
R
,
Chiu Kit
T

2002
Organochlorine exposure and age at natural menopause.
Epidemiology
13
:
729
733
33.

Akkina
JE
,
Reif
JS
,
Keefe
TJ
,
Bachand
AM

2004
Age at natural menopause and exposure to organochlorine pesticides in Hispanic women
.
J Toxicol Environ Health
,
Part A 67
34.

Yu
ML
,
Guo
YL
,
Hsu
CC
,
Rogan
WJ

2000
Menstruation and reproduction in women with polychlorinated biphenyl (PCB) poisoning: long-term follow-up interviews of the women from the Taiwan Yucheng cohort.
Int J Epidemiol
29
:
672
677
35.

Farr
SL
,
Cai
J
,
Savitz
DA
,
Sandler
DP
,
Hoppin
JA
,
Cooper
GS

2006
Pesticide exposure and timing of menopause: the Agricultural Health Study.
Am J Epidemiol
163
:
731
742
36.

Dickerson
SM
,
Gore
AC

2007
Estrogenic environmental endocrine-disrupting chemical effects on reproductive neuroendocrine function and dysfunction across the life cycle.
Rev Endocr Metab Disord
8
:
143
159
37.

Baldridge
MG
,
Stahl
RL
,
Gerstenberger
SL
,
Tripoli
V
,
Hutz
RJ

2003
Modulation of ovarian follicle maturation in Long-Evans rats exposed to polychlorinated biphenyls (PCBs) in-utero and lactationally.
Reprod Toxicol
17
:
567
573
38.

Hunt
PA
,
Koehler
KE
,
Susiarjo
M
,
Hodges
CA
,
Ilagan
A
,
Voigt
RC
,
Thomas
S
,
Thomas
BF
,
Hassold
TJ

2003
Bisphenol A exposure causes meiotic aneuploidy in the female mouse.
Curr Biol
13
:
546
553
39.

Gore
AC

2010
Neuroendocrine targets of endocrine disruptors.
Hormones
9
:
16
27
40.

Clarkson
J
,
Boon
WC
,
Simpson
ER
,
Herbison
AE

2009
Postnatal development of an estradiol-kisspeptin positive feedback mechanism implicated in puberty onset.
Endocrinology
150
:
3214
3220
41.

Roa
J
,
Aguilar
E
,
Dieguez
C
,
Pinilla
L
,
Tena-Sempere
M

2008
New frontiers in kisspeptin/GPR54 physiology as fundamental gatekeepers of reproductive function.
Front Neuroendocrinol
29
:
48
69
42.

Eghlidi
DH
,
Haley
GE
,
Noriega
NC
,
Kohama
SG
,
Urbanski
HF

2010
Influence of age and 17β-estradiol on kisspeptin, neurokinin B, and prodynorphin gene expression in the arcuate-median eminence of female rhesus macaques.
Endocrinology
151
:
3783
3794
43.

Bateman
HL
,
Patisaul
HB

2008
Disrupted female reproductive physiology following neonatal exposure to phytoestrogens or estrogen specific ligands is associated with decreased GnRH activation and kisspeptin fiber density in the hypothalamus.
Neurotoxicology
29
:
988
997
44.

Bellingham
M
,
Fowler
PA
,
Amezaga
MR
,
Rhind
SM
,
Cotinot
C
,
Mandon-Pepin
B
,
Sharpe
RM
,
Evans
NP

2009
Exposure to a complex cocktail of environmental endocrine-disrupting compounds disturbs the kisspeptin/GPR54 system in ovine hypothalamus and pituitary gland.
Environ Health Perspec
117
:
1556
1562
45.

Dickerson
SM
,
Cunningham
SL
,
Patisaul
HB
,
Woller
MJ
,
Gore
AC

2011
Endocrine disruption of brain sexual differentiation by developmental PCB exposure.
Endocrinology
152
:
581
594
46.

Oakley
AE
,
Clifton
DK
,
Steiner
RA

2009
Kisspeptin signaling in the brain.
Endocr Rev
30
:
713
743
47.

Smith
JT

2008
Kisspeptin signalling in the brain: steroid regulation in the rodent and ewe.
Brain Res Rev
57
:
288
298
48.

Mori
H
,
Matsuda
KI
,
Tsukahara
S
,
Kawata
M

2010
Intrauterine position affects estrogen receptor α expression in the ventromedial nucleus of the hypothalamus via promoter DNA methylation.
Endocrinology
151
:
5775
5781
49.

Roth
TL
,
Lubin
FD
,
Funk
AJ
,
Sweatt
JD

2009
Lasting epigenetic influence of early-life adversity on the BDNF gene.
Biol Psychiatry
65
:
760
769
50.

Ho
SM
,
Tang
WY
,
Belmonte de Frausto
J
,
Prins
GS

2006
Developmental exposure to estradiol and bisphenol A increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4.
Cancer Res
66
:
5624
5632
51.

Hernández-Hernández
JM
,
Delgado-Olguín
P
,
Aguillón-Huerta
V
,
Furlan-Magaril
M
,
Recillas-Targa
F
,
Coral-Vázquez
RM

2009
Sox9 represses α-sarcoglycan gene expression in early myogenic differentiation.
J Mol Biol
394
:
1
14
52.

Peng
H
,
Ivanov
AV
,
Oh
HJ
,
Lau
YF
,
Rauscher
FJ

2009
Epigenetic gene silencing by the SRY protein is mediated by a KRAB-O protein that recruits the KAP1 co-repressor machinery.
J Biol Chem
284
:
35670
35680
53.

Sumanasekera
WK
,
Tien
ES
,
Davis
JW
,
Turpey
R
,
Perdew
GH
,
Vanden Heuvel
JP

2003
Heat shock protein-90 (Hsp90) acts as a repressor of peroxisome proliferator-activated receptor-α (PPARα) and PPARβ activity.
Biochemistry
42
:
10726
10735
54.

Tanabe
M
,
Sasai
N
,
Nagata
K
,
Liu
XD
,
Liu
PC
,
Thiele
DJ
,
Nakai
A

1999
The mammalian HSF4 gene generates both an activator and a repressor of heat shock genes by alternative splicing.
J Biol Chem
274
:
27845
27856
55.

Singh
IS
,
He
JR
,
Calderwood
S
,
Hasday
JD

2002
A high affinity HSF-1 binding site in the 5′-untranslated region of the murine tumor necrosis factor-α gene is a transcriptional repressor.
J Biol Chem
277
:
4981
4988
56.

Numoto
M
,
Yokoro
K
,
Koshi
J

1999
ZF5, which is a Kruppel-type transcriptional repressor, requires the zinc finger domain for self-association.
Biochem Biophys Res Commun
256
:
573
578
57.

Lo
KA
,
Bauchmann
MK
,
Baumann
AP
,
Donahue
CJ
,
Thiede
MA
,
Hayes
LS
,
des Etages
SA
,
Fraenkel
E

2011
Genome-wide profiling of H3K56 acetylation and transcription factor binding sites in human adipocytes.
PLoS One
6
:
e19778
58.

Ivanova
MM
,
Luken
KH
,
Zimmer
AS
,
Lenzo
FL
,
Smith
RJ
,
Arteel
MW
,
Kollenberg
TJ
,
Mattingly
KA
,
Klinge
CM

2011
Tamoxifen increases nuclear respiratory factor 1 transcription by activating estrogen receptor β and AP-1 recruitment to adjacent promoter binding sites.
FASEB J
25
:
1402
1416
59.

Franczak
A
,
Nynca
A
,
Valdez
KE
,
Mizinga
KM
,
Petroff
BK

2006
Effects of acute and chronic exposure to the aryl hydrocarbon receptor agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin on the transition to reproductive senescence in female Sprague-Dawley rats.
Biol Reprod
74
:
125
130
60.

Palanza
P
,
Morellini
F
,
Parmigiani
S
,
vom Saal
FS

2002
Ethological methods to study the effects of maternal exposure to estrogenic endocrine disrupters: a study with methoxychlor.
Neurotoxicol Teratol
24
:
55
69
61.

ATSDR
2002
Toxicological profile for methoxychlor
.
Atlanta
:
United States Department of Health and Human Services, Public Health Service
62.

Uzumcu
M
,
Kuhn
PE
,
Marano
JE
,
Armenti
AE
,
Passantino
L

2006
Early postnatal methoxychlor exposure inhibits folliculogenesis and stimulates anti-Mullerian hormone production in the rat ovary.
J Endocrinol
191
:
549
558
63.

Anway
MD
,
Cupp
AS
,
Uzumcu
M
,
Skinner
MK

2005
Epigenetic transgenerational actions of endocrine disruptors and male fertility.
Science
308
:
1466
1469
64.

Miller
BH
,
Gore
AC

2001
Alterations in hypothalamic insulin-like growth factor-I and its associations with gonadotropin releasing hormone neurones during reproductive development and ageing.
J Neuroendocrinol
13
:
728
736
65.

Walker
DM
,
Juenger
TE
,
Gore
AC

2009
Developmental profiles of neuroendocrine gene expression in the preoptic area of male rats.
Endocrinology
150
:
2308
2316
66.

Dickerson
SM
,
Cunningham
SL
,
Gore
AC

2011
Prenatal PCBs disrupt early neuroendocrine development of the rat hypothalamus.
Toxicol Appl Pharmacol
252
:
36
46

Author notes

Present address for A.E.A.: Stony Brook University, Department of Neurosurgery, Stony Brook, New York 11794.