Skip to main content
Endocrinology logoLink to Endocrinology
. 2016 Jul 11;157(9):3588–3603. doi: 10.1210/en.2016-1087

The Methylcytosine Dioxygenase Ten-Eleven Translocase-2 (tet2) Enables Elevated GnRH Gene Expression and Maintenance of Male Reproductive Function

Joseph R Kurian 1,, Somaja Louis 1, Kim L Keen 1, Andrew Wolfe 1, Ei Terasawa 1, Jon E Levine 1
PMCID: PMC5007894  PMID: 27384303

Abstract

Reproduction depends on the establishment and maintenance of elevated GnRH neurosecretion. The elevation of primate GnRH release is accompanied by epigenetic changes. Specifically, cytosine residues within the GnRH gene promoter are actively demethylated, whereas GnRH mRNA levels and peptide release rise. Whether active DNA demethylation has an impact on GnRH neuron development and consequently reproductive function remains unknown. In this study, we investigated whether ten-eleven translocation (tet) enzymes, which initiate the process of active DNA demethylation, influence neuronal function and reproduction. We found that tet2 expression increases with age in the developing mouse preoptic area-hypothalamus and is substantially higher in a mature (GT1–7) than an immature (GN11) GnRH cell line. GnRH mRNA levels and mean GnRH peptide release elevated after overexpression of tet2 in GN11 cells, whereas CRISPR/cas9-mediated knockdown of tet2 in GT1–7 cells led to a significant decline in GnRH expression. Manipulations of tet2 expression altered tet2 genome binding and histone 3 lysine 4 trimethylation abundance at the GnRH promoter. Mice with selective disruption of tet2 in GnRH neurons (GnRH-specific tet2 knockout mice) exhibited no sign of altered pubertal timing in either sex, although plasma LH levels were significantly lower, and fecundity was altered specifically in adult male GnRH-specific tet2 knockout animals, indicating that tet2 may participate in the maintenance GnRH neuronal function. Exposure to bisphenol A, an environmental contaminant that alters GnRH neuron activity, caused a shift in tet2 subcellular localization and a decrease in histone 3 lysine 4 trimethylation abundance at the GnRH promoter. Finally, evaluation of tet2 protein interactions in GT1–7 cells suggests that the influence of tet2 on neuronal function are not limited to nuclear mechanisms but could depend on mitochondrial function, and RNA metabolism. Together, these studies implicate tet2 in the maintenance of GnRH neuronal function and neuroendocrine control of male reproduction.


Reproductive function depends on the establishment and maintenance of elevated GnRH neurosecretion. We previously discovered that elevated GnRH release coincides with an epigenetic process during neuronal development. Specifically, cytosine methylation status across a distal 5′ portion of the rhesus monkey GnRH gene declines as GnRH peptide release begins to rise during in vitro development of GnRH neurons (1). We subsequently found that a similar demethylation phenomenon occurs across pubertal development in male rhesus monkeys in vivo (2), suggesting developmental epigenetic modifications are not static at the GnRH gene. The dynamicity of GnRH epigenetic regulation (histone modifications) has been reported (3) in a region that 1) controls mouse pubertal timing and periodic reproductive cycling (4) and 2) is analogous to the primate genome location subject to developmental active DNA demethylation (1, 2). Together, this highlights the likely role of epigenetic regulation of GnRH in the context of puberty, but perhaps more importantly, the regulation of age associated declines in hypothalamic GnRH mRNA content and reproductive physiology (5, 6). To date, the enzymes responsible for epigenetic manipulation of GnRH gene expression are not defined.

Recently, a family of ten-eleven translocation (tet) enzymes (tet1, tet2, and tet3) was found to initiate the process of active DNA demethylation. Tet enzymes oxidize 5-methylcytosine to form 5-hydroxymethylcytosine (5hmC) and further oxidize 5hmC to generate 5-formylcytosine and 5-carboxylcytosine (79). 5-Formylcytosine and 5-carboxylcytosine are subsequently recognized and removed by base excision repair to complete the process of active DNA demethylation (10, 11). Importantly, 5hmC is not solely an intermediate metabolite of active demethylation. Some gene promoters, enhancers, and exons, are enriched for 5hmC (1219), and binding of 5hmC by the MBD3/NURD complex and MeCP2 further shapes chromatin structure (2022). In addition, an interaction between tet2 and the SET1/COMPASS complex promotes histone 3 lysine 4 trimethylation (H3K4me3) (23). The promotion of H3K4me3 is particularly intriguing in the context of GnRH neuron secretory status, because the abundance of this permissive histone modification is significantly higher at the GnRH gene in differentiated compared with undifferentiated GnRH neuronal cell lines (3).

Based on their control of DNA methylation dynamics and promotion of H3K4me3, we tested the hypotheses that tet enzymes modulate GnRH expression and secretion and that tet activity in GnRH neurons modifies the timing of puberty and/or reproductive function. To test these hypotheses, we first evaluated the expression of tet enzymes in GnRH cell lines and tet2 in the developing preoptic area and mediobasal hypothalamic (POA-MBH) tissues of male and female mice. We subsequently assessed the impact of tet2 activity on GnRH gene expression and chromatin modifications. Finally, we generated mice with specific ablation of tet2 activity in GnRH neurons to evaluate the role of tet2 in puberty and adult reproductive function.

Materials and Methods

Animals

Mice were housed under a 12-hour light, 12-hour dark cycle and had access to water and chow (Harlan/Teklad 2019) ad libitum. All experimental procedures adhered to guidelines provided in the National Institutes of Health Guide for the Care and Use of Laboratory Animals and were approved by the Institutional Animal Care and Use Committee at the University of Wisconsin-Madison.

Cell culture

Growth conditions

GN11 cells (kindly provided by Dr Sally Radovick, Rutgers University) were grown in DMEM (Sigma-Aldrich) supplemented with 10% fetal bovine serum stripped with dextran-coated charcoal to remove steroids and other small molecules (HyClone), 25mM glucose, 5mM L-glutamine, 100-U/mL penicillin, and 100-μg/mL streptomycin (Gibco). This medium is referred to as “complete medium.” GT1–7 cells (kindly provided by Dr Pamela Mellon, University of California, San Diego, CA) were grown in a similar medium, although the supplemental serum was heat inactivated. Cultures were maintained at 5% CO2 and 37°C. Cells used for perifusion experiments were grown on plastic coverslips placed in 6-well plates, all other cultures were grown directly in 6-well or 150-mm plates.

Tet2 overexpression

For overexpression of tet2, GN11 cells were transferred to serum and antibiotic-free medium and transiently transfected with a pCMV6-entry expression plasmid containing the mouse tet2 open reading frame (catalog number MR227061; OriGene) using FuGENE HD Transfection Reagent (Roche Diagnostics) at 37°C, following the manufacturer's instructions. Tet2 expression was confirmed by qPCR and Western blotting. Control (CON) GN11 cultures were transfected with the same plasmid containing no insert (catalog number PS100001; OriGene). Medium was aspirated and replaced with complete medium 24 hours after transfection. Experiments were conducted 48 hours after transfection. In CON cultures, cells were harvested, pelleted, and kept at −80°C until RNA extraction.

Tet2 gene disruption

GT1–7 cells were seeded in 6-well tissue culture plates (2.5 × 103/well) and grown to 80% confluency in antibiotic-free normal growth media containing fetal bovine serum. Knockdown was performed with the Santa Cruz Biotechnology, Inc CRISPR/Cas9 tet2 gene disruption materials. Briefly, a tet2-specific or CON pool of CRISPR/Cas9 knockout (KO) plasmids (catalog number sc-431916) were cotransfected with tet2 HDR plasmids (catalog number sc-431916-HDR); the latter plasmids allowed for puromycin selection of KO plasmid-transfected cells. Transfections used 3 μg of each plasmid and FuGENE HD Transfection Reagent according to manufacturer recommendations. After puromycin selection, cells were cultured in normal growth media and allowed to reach 90% confluency before subsequent assays, including evaluation of gene silencing, which was confirmed by Western blotting and qPCR.

Quantitative real-time PCR

Total RNA was extracted from pelleted cells or POA-MBH tissue using TRIzol reagent (Invitrogen) according to the manufacturer's protocol. POA-MBH tissues were dissected using the following confines. Anterior, the anterior limit of the nucleus of the vertical limb of the diagonal band (bregma, +1.34 mm); posterior, the posterior limit of the mammillary nucleus (bregma, −3.40 mm); lateral, the external borders of the medial POA (rostral) and mammillary nucleus (caudal); and dorsal, 1.5 mm in depth. Two micrograms of RNA were reverse transcribed using the StrataScript first-strand synthesis system kit (Stratagene). Real-time quantitative PCR was performed in triplicate using SyberGreen MasterMix (Bio-Rad) and the ICycler quantitative PCR machine (Bio-Rad). 18S RNA was used as internal control for cDNA input; 18S critical threshold (Ct) values were stable within each analysis between conditions. Primer sets are listed in Supplemental Table 1. PCR efficiency was determined with a 10-fold serial dilution of cDNA as previously described (24). PCR conditions were optimized to generate more than 95% and less than 105% efficiency. Fold changes in relative gene expression were calculated by 2−ΔCt or 2−Δ(ΔCt) according to Livak and Schmittgen (25). Briefly, for each sample, a ΔCt was calculated to normalize for the internal control (18S) using the equation: ΔCt = Ct(gene) − Ct(18S). For comparisons without a specified CON sample for comparison, expression levels were calculated as 2−ΔCt. To obtain differences between experimental and CON conditions (ie, GN11 cells), ΔΔCt was calculated: ΔCt(sample) − ΔCt(GN11). Relative mRNA levels were then calculated using the equation fold difference = 2−ΔΔCt, with GN11 relative expression (ΔCt(GN11) − ΔCt(GN11)) consequently set to 1.

Chromatin immunoprecipitation

Enrichment of tet2, H3K4me3, and H3K27me3 was measured at 4 regulatory regions of the mouse GnRH gene (previously described in Ref. 3) in GN11, GT1–7, GN11 cells overexpressing tet2, and GT1–7 cells with crispr/cas9-mediated tet2 gene disruption. Forty-eight hours after transfection, 4 cultures of each condition (confluent 150-mm plates) were treated with 1% formaldahyde for 10 minutes at room temperature. Cell lysis, sonication (4 × 30-s pulses on ice in a Fisher Scientific 550 Sonic Dismembranator set at level 4 to shear DNA into 300- to 600-bp segments), immunoprecipitation and DNA purification were carried out according to manufacturer protocol (MAGNA ChIP A/G; catalog number 17–10085; Millipore) Briefly, lysates were prepared by exposure of cells to ice cold lysis buffer for 20 minutes, followed by gentle disruption through shaking and repeated pipetting up and down. The antibodies were covalently cross linked to the magnetic beads with disuccinimidyl suberate, then cleared lysates were incubated with the beads while rolling at 4°C overnight. Beads were then captured and washed using a magnetic stand. Immunoprecipitated protein were then eluted and analyzed by Western blotting to determine enrichment of tet2. Eluted protein were sent to MS Bioworks for in gel digestion followed by mass spectrometry and bioinformatic analysis/identification of peptides measured in each sample. Tet2-binding partners were considered proteins with at least 4 unique peptides detected and no less than 3 times the unique peptides detected in IgG-precipitated eluates. Antibody information for all methods is presented in Table 1.

Table 1.

Antibodies Used in These Studies

Peptide/Protein Target Antigen Sequence Name of Antibody Manufacturer, Catalog Number, and/or Name of Individual Providing the Antibody Species Raised (Monoclonal or Polyclonal) Dilution/Amount (Purpose, if Multiple)
Tet2 Peptide mapping within an internal region of TET2 of human origin Tet2 (S-13) Santa Cruz Biotechnology, Inc, sc-136926 Rabbit (polyclonal) 1:1000 (Western)
1:1000 (IHC)
4 μg (IP)
H3K4me3 Synthetic peptide corresponding to residues surrounding and including trimethlyated Lys 4 of histone H3 Antitrimethyl-histone H3 (Lys4) Millipore, 07-473 Rabbit (polyclonal) 2 μg
H3K27me3 KLH-conjugated, synthetic 2X-branched peptide containing the sequence … AR (me3K)SAP …, in which me3K corresponds to trimethyl-lysine at residue 27 of human histone H3 Antitrimethyl-histone H3 (Lys27) Millipore, 07-479 Rabbit (polyclonal) 2 μg
GnRH Synthetic peptide that represents the full-length GnRH I decapeptide of rat origin HU11B Dr Henryk Urbanski (Oregon National Primate Research Center) Mouse (monoclonal) 1:1000 (IHC)
GnRH Synthetic LHRH LRH13 Dr M. K. Park (University of Tokyo, Japan) Mouse (monoclonal) 1:2500 (IHC)
GnRH Mammalian GnRH R1245 Dr Terry M. Nett (Colorado State University, Fort Collins) CO) Rabbit (polyclonal) 1:420 (RIA)
LH Cynomolgus LH-1-1 (AFP-6936A) AFP-342994 National Hormone and Peptide Program (Torrance, CA) Rabbit (polyclonal) 1:3000

Coimmunoprecipitation

GT1–7 cells were cultured to full confluence in 150-mm dishes. Cells from 2 plates were combined for each immunoprecipitation; 2 independent immunoprecipitations were conducted for each antibody: tet2 (catalog number sc-136926; Santa Cruz Biotechnology, Inc) and normal rabbit IgG (catalog number 12–370; Millipore). Co-IP were performed using the Pierce Crosslink Magnetic IP/Co-IP kit (catalog number 88805) according to manufacturer recommendations. Two micrograms of each antibody (tet2 [sc-136926; Santa Cruz Biotechnology, Inc], H3K4me3 [07–473; Millipore], H3K27me3 [07–449; Millipore], and normal rabbit IgG [12–370; Millipore]) were used for each overnight immunoprecipitation at 4°C. To measure fold enrichment of each histone modification and tet2 binding, quantitative PCR was performed with 2 ng of purified DNA, ABI SYBR green PCR master mix (catalog number 4309155; ABI) and primers listed in Supplemental Table 1. Reactions were run in triplicate using an ABI 7300 system 2 minutes at 50°C, 10 minutes at 94°C then 45 cycles of 15 seconds at 94°C then 1 minute at 60°C followed by a melt curve analysis. Average Ct values were calculated for each primer set and fold enrichment was calculated relative to IgG (2 (average IgG Ct − average-specific antibody Ct), data are presented as mean of 4-fold enrichment values ± SEM.

Generation of GnRH-cre/tet2 lox/lox (gTKO) mice

To specifically ablate tet2 activity in GnRH neurons, a mouse strain was used in which exon 3 of the tet2 gene, which encodes greater than half of the murine protein and is included in the 2 described tet2 transcripts, is flanked by loxP sites (tet2lox/lox). Deletion of exon 3 using this model results in a lack of tet2 transcript in Cre-expressing cells/tissues (26). These mixed B6/129S background mice were backcrossed with a pure C57BL/6J background strain. For GnRH neuron-specific tet2 ablation, GnRHcre male mice (27) were crossed with female tet2lox/lox mice, and then the resultant GnRH cre+/tet2lox/wt males were crossed with tet2lox/lox female mice. The F2 progeny included GnRHcre+/tet2lox/lox (gTKO) mice and GnRHcre/tet2lox/lox mice. The latter were used as CON animals. Genotyping. Toe samples were clipped from mice 1–2 weeks after birth and subsequently incubated in lysis buffer containing proteinase K at 95°C for 3 hours followed by the addition of KCl and centrifugation to pellet cell/tissue debris. The DNA pellet was washed in 70% ethanol and repelleted. DNA pellets dissolved in water were used for PCR analysis using standard conditions. Primer sequences are listed in Supplemental Table 1.

Immunohistochemistry

On brain tissue

Two (1 male and 1 female) mice for each condition (CON and gTKO) were perfused transcardially with 4% paraformaldehyde (fixative) under ketamine/xylazine anesthesia. Brains were removed and soaked in fixative for an additional 24 hours, then transferred to 30% sucrose for 72 hours. Brains were then frozen in the tissue freezing medium OCT (Leica) and cut into 14-μm sections with a cryostat. Coronal brain sections were blocked in 1× PBS, 0.1% Triton X-100, 5% normal goat serum, washed twice with PBS blocked again with mouse IgG for 1 hour. After 2 washes, sections were exposed to rabbit polyclonal Tet2 (S-13) antibody (1:1000 dilution; catalog number sc-136926; Santa Cruz Biotechnology, Inc) for 2 days at 4°C. The next day, after washes, sections were treated with goat antirabbit IgG Alexa Fluor 594 (1:1000 dilution; Abcam) to complete tet2 staining. Subsequently, sections washed with PBS, blocked with 5% normal goat serum, then exposed to GnRH primary antibody HU11B (1:1000 dilution; a gift from Henryk Urbanski, Oregon National Primate Research Center) or LRH13 (1:2500 dilution, a gift from Dr M. K. Park, University of Tokyo, Japan) for 2 days at 4°C. On the final day, sections were washed then exposed to goat antimouse IgM Alexa Fluor 488 (1:1000 dilution; Abcam). Sections were then incubated with DAP1 for nuclear staining and coverslipped with Vectashield (Vector Laboratories). Colocalization of Tet2 in GnRH neurons was examined by confocal microscopy (Nikon A1R Ti-E), and quantitative analysis was conducted with fluorescent microscopy (Nikon, Microphot FXA).

In cultured cells

GT1–7 cells were grown on Lab-Tek chamber slides. After treatment with 10nM bisphenol A (BPA) (catalog number 239658, Sigma-Aldrich) for 2 hours, the cells were fixed with 4% methanol-free formaldehyde. After blocking the slides with normal blocking serum (5% goat serum), the cells were incubated with primary antibody rabbit polyclonal Tet2 (S-13) (1:1000 dilution, catalog number sc-136926; Santa Cruz Biotechnology, Inc) overnight at 4°C. After 3 washes with 1× PBS, the cells were incubated with goat antirabbit IgG secondary antibody-Alexa Flour 488 conjugate (2 μg/μL, catalog number A11008; Thermo Fisher Scientific) in 1× PBS with 1% goat serum for 1 hour in the dark. After washing, the slides were mounted with Vectashield Hard set mounting medium with DAP1 (Vector Laboratories).

Perifusion experiments

Methods have been previously described in detail (28). Briefly, cultures containing GnRH cells were mounted in Sykes-Moore chambers as described by Martínez de la Escalera et al (29), ie, 2 coverslips were placed, face to face, separated by a rubber O-ring, forming a chamber with a volume of 200 μL. Cells were then perifused with artificial cerebrospinal fluid containing 0.1% glucose (pH 7.4) under 95% O2 and 5% CO2 at 37°C. Perifusates were collected at 25 μL/min in 20-minute fractions for 6 hours using the ACUSYST (Endotronics) perifusion system. To test the viability of cells, 56-mm K+ was perifused for 20 minutes during the last hour (h 6) of the experiment. Samples were stored at −80°C until they were assayed for GnRH by RIA.

Radioimmunoassays

LH measurements:

Animals were anesthetized with isoflurane and blood was withdrawn by cardiac puncture between 9 and 10 am. Duplicate 50-μL serum samples were assayed for LH using RIA reagents obtained from the National Institute of Diabetes and Digestive and Kidney Diseases, including the rat LH standard (RP-3) and S-11 antibody; the iodinate was prepared by the Assay Services Core Unit at the Wisconsin National Primate Research Center. The assay had a lower limit of detection of 0.028 ng/mL at 95% binding. One assay was performed with an intraassay coefficient of variation (n = 158) 7.1%.

GnRH measurements

GnRH concentrations from perifusion experiments were measured in duplicate 200-μL samples with a RIA using antiserum R-1245 (a gift from Dr T. Nett, Colorado State University, Fort Collins, CO) as described previously (30). Synthetic GnRH (Richelieu Laboratory, Inc) was used for both the trace and the reference standard. The sensitivity of the assay, at 95% binding, was 0.05 pg/tube. All samples were run in the same assay; the intraassay coefficient of variation (n = 108) was 8.4%.

Western immunoblotting

Cells were grown to 90% confluency in tissue culture flasks. Cell lysates were prepared by sonication and collected after centrifugation of cellular debris. Protein concentrations were measured using Pierce bicinchoninic acid assay kit (catalog number 23227). Proteins were separated using a 4%–15% SDS-PAGE gel (Bio-Rad) after equal loading of 15 μg of protein per well. Proteins were transferred to PVDF membrane-Immobilin-FL (Millipore) and probed overnight at 4°C with the primary antibody rabbit polyclonal tet2 (S-13) (1:100 dilution, catalog number sc-136926; Santa Cruz Biotechnology, Inc; tet2 detected at molecular weights 133 and 131) or mouse anti-β-actin antibody (1:2000; Sigma-Aldrich) after completion of the initial tet2 western followed by membrane stripping. After 4 washes with 1× PBS+0.1% Tween 20, the membranes were treated with secondary antibody IRDye 800CW goat antirabbit (1:5000) or IRDye680LT goat antimouse (1:10 000) for 1 hour at room temperature with gentle shaking. The blots were analyzed using the LI-COR Odyssey Infrared Imaging system. Quantification of the relative band densities (tet2/β-actin) was completed using ImageJ on 2 separate blots.

Vaginal opening (V.O.) and estrous cyclicity

Beginning at 20 days of age, females were examined for the onset of V.O. To examine the possible effects of genotype on estrous cyclicity, vaginal lavages from female mice (beginning at V.O. through approximately 55 d of age) were obtained and viewed under a microscope daily (9–10 am) for at least 21 consecutive days. Estrous stages were defined based on the primary cell types present in vaginal cytology samples as follows: leukocytic (L), metestrus/diestrus; nucleated (N), proestrus; and cornified (C), estrus. First estrus was determined as the first complete cycle of the pattern proestrus-estrus-2 consecutive days of metestrus/diestrus, as described previously (see reference 43 below).

Body weight measurements

Body weights were recorded daily (9–11 am).

Reproductive function (fecundity)

At p60, each mouse was paired with 1 CON animal of the opposite sex of proven fertility. When females were visibly pregnant, animals were separated and the number of days from initial pairing to the birth of a litter was recorded. The total number of pups per litter was also recorded.

Data analyses and statistics

Data are presented as the mean ± SEM. One-way ANOVA followed by Tukey post hoc was used to determine statistical significance related to tet2 overexpression and knockdown and GN11 cell stimulation (see figures 3 and 4 below). Two-way ANOVA followed by Tukey post hoc tests were used to determine statistical significance in chromatin-immunoprecipitation (ChIP) experiments and mouse hypothalamic mRNA content. Difference in the colocalization rate in CON and gTKO brains was tested with χ2 test. Two-tailed, unpaired t tests were used to determine statistical significance for the remaining comparisons. Differences were considered significant when P < .05. Statistics were performed using GraphPad Prism software, version 6.0e.

Results

Developmental expression of tet enzymes in vitro

To examine whether the process of GnRH neuronal differentiation requires active DNA demethylation, we hypothesized that tet enzyme expression or activity increases during the course of development. We first evaluated relative tet1, tet2, and tet3 gene expression in 2 GnRH-secreting cell lines; GN11 and GT1–7 cells. This comparison capitalizes on the distinct stages of development between these cells. GN11 cells, originally isolated from the mouse nasal region (31), express GnRH at very low levels and represent GnRH neurons at the earliest stages of differentiation, whereas GT1–7 cells, which were isolated from the adult mouse hypothalamus (32), are characterized by mature activity patterns, including elevated GnRH mRNA levels. We found that tet1 is expressed in both cell lines, with a trend toward higher expression in GT1–7 cells (n = 3 culture for each cell line; GN11, 0.536 ± 0.077 vs GT1–7, 0.858 ± 0.126; P = .09) (Figure 1). Tet3 is also expressed in both cell lines with no difference in expression (n = 3 cultures for each cell line; GN11, 0.203 ± 0.046 vs GT1–7, 2.15 ± 0.050) (Figure 1). Tet2 expression was scarcely detectable in the immature cells but strongly expressed in the mature cells (n = 3 cultures for each cell line; GN11, 0.020 ± 0.008 vs GT1–7, 0.648 ± 0.165; P = .02) (Figure 1).

Figure 1.

Figure 1.

Tet enzyme expression in GnRH cell lines. Comparison of tet enzyme expression in undifferentiated (GN11) and differentiated (GT1–7) GnRH cell lines. Data are presented as mean ± SEM; t tests compared means between cell lines for each tet enzyme; *, P = .02.

Developmental expression of tet2 in vivo

Using quantitative PCR and total RNA isolated from POA-MBH tissues, we also found that tet2 expression increases in male and female mice between postnatal day (p)15 and p25 (n = 5 for each sex and time point; male p15, 0.442 ± 0.048 vs p25, 0.822 ± 0.10; t test, P = .007; and female p15, 0.190 ± 0.03 vs p25, 1.117 ± 0.39; t test, P = .047) (Figure 2). Sex differences in tet2 expression were not observed (two-way ANOVA, sex F1,16 = 0.0112, P = .9171; age F1,16 = 10.33, P = .0054, Tukey post hoc test at α = 0.05: p15 female < p25 female).

Figure 2.

Figure 2.

Tet2 expression in the medial basal hypothalamus. Comparison of tet2 expression in male and female medial basal hypothalamus on p15 and p25. Data are presented as mean ± SEM; t tests compared means between postnatal days for each sex; **, P = .007; *, P = .047. No sex differences in tet2 expression were observed (two-way ANOVA, sex F1,16 = 0.0112, P = .9171; age F1,16 = 10.33, P = .0054, Tukey post hoc test at α = 0.05: p15 female < p25 female).

Tet2 activity in GnRH neurons in vitro

We reasoned that if GnRH gene expression is the consequence of the elevated levels of tet2 in GnRH cell lines, then GnRH mRNA levels should increase when tet2 is overexpressed in GN11 cells; conversely, GnRH mRNA levels might decline after disruption of tet2 expression in GT1–7 cells. We transfected GN11 cells with either a plasmid that encodes murine tet2 or a CON plasmid (GN11+tet2); GT1–7 cells were either cotransfected with a plasmid that encoded tet2 gene-specific guide RNAs and cas9 and a plasmid encoding selection genes (GT1–7(−)tet2) or with the CON CRISPR/Cas9 plasmid alone (CON GT1–7). Tet2 mRNA expression was significantly higher in tet2-transfected GN11 and GT1–7 cells compared with CON-transfected GN11 and GT1–7(−)tet2 cells (n = 3 cultures for each condition, GN11: 1, GT1–7: 11.21 ± 1.8, GT1–7(−)tet2: 0.515 ± 0.10, GN11+tet2: 15.95 ± 3.19; ANOVA F3,8 = 17.37, P = .0007; Tukey post hoc at α = 0.05: GT1–7 and GN11+tet2 > GN11, GT1–7 and GN11+tet2 > GT1–7(−)tet2, GT1–7 = GN11+tet, GN11 = GT1–7(−)tet2) (Figure 3A). Expression differences were confirmed by Western blotting (Figure 3A, inset) followed by quantification of tet2/β-actin band intensity ratios using ImageJ (n = 2 blots, GN11: 0.193 ± 0.02, GT1–7: 0.490 ± 0.04, GT1–7(−)tet2: 0.205 ± 0.06, GN11+tet2: 0.563 ± 0.06; ANOVA F3,4 = 15.67, P = .011; Tukey post hoc at α = 0.05: GT1–7 and GN11+tet2 > GN11, GT1–7 and GN11+tet2 > GT1–7(−)tet2, GT1–7 = GN11+tet, GN11 = GT1–7(−)tet2). GnRH mRNA levels rose after tet2 overexpression in GN11 cells (n = 3 cultures for each condition, GN11: 0.110 ± 0.02, GN11+tet2: 1.03 ± 0.29; t test, P = .034) (Figure 3B), whereas CRISPR/cas9-mediated disruption of GT1–7 cell tet2 expression caused a significant decrease in GnRH mRNA levels (n = 4 cultures per condition, GT1–7: 3.11 ± 0.069, GT1–7(−)tet2: 0.79 ± 0.12; t test: P < .001) (Figure 3C), indicating that tet2 not only enables GnRH expression in immature cells but also has substantial influence over the maintenance of GnRH mRNA level in mature cells. Notably, using a perifusion method we found that the rise in GN11 cell GnRH mRNA after tet2 overexpression had an apparent effect on peptide release, as GN11+tet2 cells released more GnRH peptide compared with CON-transfected cells (n = 3 cultures per condition; GN11: 2.10 ± 0.26 vs GN11+tet2: 2.67 ± 0.12 pg/mL per 10 minutes (P = .043) (Figure 3D).

Figure 3.

Figure 3.

Tet2 drives GnRH gene expression and peptide release in undifferentiated GnRH neurons while maintaining elevated GnRH mRNA levels in differentiated GnRH neurons. A, Comparison of tet2 mRNA in undifferentiated (GN11), differentiated (GT1–7), GT1–7 cells with disrupted tet2 expression (GT1–7(−)tet2), and GN11 cells overexpressing tet2 (GN11+tet2). Data are presented as mean ± SEM, ANOVA followed by Tukey post hoc, bars with same letter are statistically equivalent; P < .001. A, inset, Western blotting indicating differences in tet2 protein levels between respective cell lines. B, Comparison of GnRH mRNA in GN11, and GN11+tet2 cell lines (t test, P = .034). C, Comparison of GnRH mRNA in GT1–7, and GT1–7(−)tet2 cell lines (t test, P < .001). D, Measurements of average GnRH peptide release from GN11 and GN11 (+) tet2. Data are presented as mean ± SEM; t test compares means; *, P = .043.

Tet2 binding and histone modifications in GnRH cell lines

In addition to, and possibly as a consequence of manipulating DNA methylation dynamics, tet2 promotes H3K4me3 (see reference 56 below), an activating histone modification. Consequently, using chromatin immunoprecipitation followed by quantitative PCR, we measured levels of H3K4me3, H3K27me3 (a restrictive histone modification), and tet2 binding across 3 enhancer regions and the promoter of the GnRH gene (Figure 4A) in GN11, GT1–7, GN11+tet2, and GT1–7(−)tet2 (n = 4 cultures for each cell line). Tet2 binding was low in GN11 cells but significantly higher in GT1–7 and GN11+tet2 cells, where binding was most pronounced at enhancer E1 and the promoter; tet2 binding significantly declined at those regions in GT1–7(−)tet2 cells (two-way ANOVA, cell line F3,55 = 39.44, P < .001; gene region F4,55 = 56.66, P < .001) (Figure 4B). H3K4me3 levels were also low in GN11 cells and higher in GT1–7 and GN11+tet2 cells. H3K4me3 was most pronounced at the promoter and to a lesser extent at E1 in GT1–7; H3K4me3 levels also increased in GN11 cells after tet2 overexpression at E1 and the promoter. On the other hand, GT1–7(−)tet2 cells exhibited significantly reduced H3K4me3 abundance at the promoter (two-way ANOVA, cell line F3,55 = 18.91, P < .001; gene region F4,45 = 32.53, P < .001) (Figure 4C). H3K27me3 levels were similar between the cell lines with some evidence of greater enrichment near E1 and the promoter (two-way ANOVA, cell line F3,55 = 1.468, P = .1316; gene region F4,45 = 7.775, P < .001) (Figure 4D).

Figure 4.

Figure 4.

Tet2 binding and chromatin modifications across the mouse GnRH gene. A, Schematic diagram (adapted from Iyer et al [3]) of the mouse GnRH gene depicting locations of enhancers E3, E2, E1, and the GnRH promoter (P). Opposing arrows show ChIP assay primer binding locations. For reference, fold enrichment was also measured at the insulin promoter (not depicted). B–D, Fixed chromatin from GN11, GT1–7, and GN11+tet2 and GT1–7(−)tet2 cell lines was sonicated then immunoprecipitated with antibodies against Tet2, H3K4me3, or H3K27me3. DNA was then analyzed by qPCR. Data are presented as fold enrichment relative to IgG (2 (IgG Ct-specific antibody Ct), mean of 4 cultures per cell line ± SEM. Regions not assigned the same letter are significantly different by Tukey post hoc; P < .05 after two-way ANOVA.

Tet2 disruption in GnRH neurons

Using conditional gene targeting, we next generated mice with specific ablation of tet2 activity in GnRH (GnRH cre X tet2 fl/fl) expressing neurons. All comparisons were between transgenic animals (gTKO: GnRH-Cre positive, tet2 fl/fl) and CON littermates (GnRH-Cre negative, Tet2 fl/fl). Double immunohistochemical staining indicated that tet2 was observed in the cytosol of CON GnRH neurons but not in gTKO neurons (Figure 5). Importantly, adjacent non-GnRH cells had tet2 expression in both CON and gTKO animals. Quantitative analysis in gTKO and CON brains indicated significantly fewer tet2-positive GnRH neurons in gTKO mice compared with CON mice in both sexes (P < .0001) (Table 2). We then evaluated tet2 and GnRH mRNA levels in the MBH/POA of male and female animals at p15, p25, and p100. Tet2 expression increased with age in both sexes across genotypes (female: two-way ANOVA; age, F2,22 = 18.33, P = .001; Tukey post hoc test at α = 0.05, p15 CON < p25 CON and p15 kTKO < p25 CON; male: two-way ANOVA, age F2,21 = 3.620, P = .045) (Figure 6, A and B). Genotype had no impact on tet2 expression in either sex (female: two-way ANOVA; age, F2,33 = 0.466, P = .632; male: two-way ANOVA; age, F2,31 = 0.1595, P = .853) (Figure 6, A and B), likely due to the maintenance of tet2 expression in non-GnRH expressing cells. An effect of genotype was evident for GnRH expression in male gTKO animals. GnRH expression was lower in gTKO males compared with CON littermates (male: two-way ANOVA; genotype, F1,23 = 6.798, P = .0157; Tukey post hoc test at α = 0.05, p15, p25, and p100 CON > p100 gTKO; female: two-way ANOVA; genotype, F1,21 = 0.3569, P = .5566) (Figure 6, C and D), with a trend toward declining GnRH expression with age across genotypes for both sexes (male: two-way ANOVA; age, F2,23 = 3.152, P = .0617; female: two-way ANOVA; age, F2,21 = 3.308, P = .0564) (Figure 6, C and D).

Figure 5.

Figure 5.

Double immunohistochemical staining for Tet2 and GnRH in the POA of GnRH cell-specific tet2 knockout mice. Photomicrographs with a confocal microscope of CON brain (top panels) and GnRH-specific tet2 knockout (gTKO) brain (bottom panels) using antibodies specific to GnRH (green, left panels) and tet2 (red, middle panels) shows that Cre-mediated recombination of the tet2 gene is restricted to GnRH neurons (overlay, right panels). Blue is a nuclear staining (DAPI). In CON animals, arrows point to a GnRH neuron with immuno-positive tet2 staining in the GnRH neuron. Arrows in gTKO panels indicate a GnRH neuron, in which tet2 staining is absent. Note that adjacent non-GnRH cells are tet2 immuno-positive.

Table 2.

Tet2 Immuno-Positive GnRH Neurons in CON and gTKO Male and Female Mice

CON
gTKO
GnRH Neuron Number Tet2 Positive GnRH Neurons Percent Positive GnRH Neuron Number Tet2 Positive GnRH Neurons Percent Positive
Male 49 46 95.6 49 4 7.7a
Female 40 38 95 45 3 6.7b

Cell count was made in 24-tissue sections in each animal.

a

χ2 = 72.03, P < .0001.

b

χ2 = 66.18, P < .0001.

Figure 6.

Figure 6.

Gene expression in the hypothalamus of GnRH cell-specific tet2 knockout mice. Expression of tet2 and GnRH in the MBH/POA of male and female CON and GnRH cell-specific tet2 knockout mice (gTKO) across development. A and B, Tet2 expression increased with age in both females (**, two-way ANOVA; age, F2,33 = 8.625, P = .001) and males (***, two-way ANOVA; age, F2,31 = 3.668, P = .0372). C and D, GnRH expression was lower in gTKO males compared with CON littermates (#, two-way ANOVA; genotype, F1,23 = 6.798, P = .0157) with a trend toward declining GnRH expression with age across genotypes for both sexes (male: two-way ANOVA; age, F2,23 = 3.152, P = .0617; female: two-way ANOVA, age, F2,21 = 3.308, P = .0564). Data are presented as expression relative to 18s; mean ± SEM, n ≥ 4 animals per sex and genotype, run in triplicate for each gene and developmental age.

Role of Tet2 in GnRH neurons in vivo

If tet2 is necessary for the maturation of GnRH neurons, then disruption of tet2 activity in GnRH neurons should influence reproductive function. We measured body growth, plasma LH (an indirect measure of GnRH neuronal activity) at p30 and p60, and early adulthood fecundity in both sexes. The day of preputial separation was evaluated in males and V.O., vaginal smears for 28 days after V.O., and a first estrus were determined for females. All comparisons were between transgenic (gTKO; GnRH-Cre positive, Tet2 fl/fl) animals and CON littermates (GnRH-Cre negative, Tet2 fl/fl). We did not find any indication of abnormal timing or progression of puberty in either sex. The average age of V.O. (CON [n = 5]: 29.0 ± 0.3, gTKO [n = 7]: 29.6 ± 0.2 days of age; t test, P = .11) (Figure 7A) and first estrus (CON [n = 5]: 34.6 ± 1.03, gTKO [n = 7]: 36.3 ± 1.56 days of age; t test, P = .43) (Figure 7A) were similar and there were no differences in percentage of time spent in each estrus stage between CON and gTKO animals (Figure 7B). Preputial separation was also similar between gTKO animals and CON littermates (CON [n = 4]: 33.0 ± 0.71, gTKO [n = 3]: 33.7 ± 1.85; t test, P = .72) (Figure 7A). Plasma LH levels exhibited typical developmental patterns and were similar for both genotypes and both sexes at p30 (female CON [n = 4]: 0.933 ± 0.24, gTKO [n = 4]: 1.54 ± 0.82 ng/mL; male CON [n = 5]: 1.90 ± 0.33, gTKO [n = 4]: 0.94 ± 0.25) (Figure 7C). There was no difference between CON and gTKO female plasma LH levels at p60 (CON [n = 3]: 0.268 ± 0.10, gTKO [n = 4]: 0.159 ± 0.014 ng/mL) (Figure 7C) or young adult female fecundity (CON [n = 5]: 23 ± 0.32 vs gTKO [n = 5]: 23.4 ± 0.45 days between pairing and birth of litter; CON [n = 5]: 7.2 ± 0.37 vs gTKO [n = 5]: 7.0 ± 0.80 pups per litter) (Figure 7D).

Figure 7.

Figure 7.

Mice with specific ablation of tet2 activity in GnRH neurons (gTKO) exhibit typical pubertal development in both sexes and male-specific deficits in gonadotropin release and reproductive function as young adults. A, The average day of v V.O., first estrus, and preputial separation is not different between gTKO and CON littermates. B, gTKO animals exhibit normal estrous cyclicity, with the percent time spent in each phase of the estrous cycle similar to CON animals. C, Plasma LH levels at p30 are similar between gTKO and CON animals of each sex. Plasma LH levels at p60 are similar between genotypes in female but lower in gTKO compared with CON males; data are presented as mean ± SEM; t tests compared means between postnatal days for each sex; *, P = .04. D, gTKO female fecundity did not appear compromised, but male gTKO took longer than CON animals to impregnate a fertile female; data are presented as mean ± SEM; t tests compared means between genotypes for each measurement; *, P = .034.

In contrast, in young adult gTKO males, plasma LH levels were significantly lower (male CON [n = 4]: 0.338 ± 0.095, gTKO [n = 4]: 0.101 ± 0.03 ng/mL; t test, P = .04) (Figure 7C) and young adult male gTKO fecundity was compromised, with gTKO males taking longer than CON littermates to impregnate a fertile female (CON [n = 4]: 23 ± 0.45 vs gTKO [n = 5]: 25.8 ± 0.85 d between pairing and birth of litter; t test, P = .034) (Figure 7D). There was no difference in number of pups per litter sired by CON or gTKO animals (CON [n = 4]: 7.60 ± 0.51 vs gTKO [n = 5]: 7.75 ± 0.25) (Figure 7D). There were also no differences in body weight observed between genotypes within sex through p100 (data not shown).

Influence of BPA on Tet2 subcellular localization and GnRH gene chromatin environment

Because in vivo data suggests that tet2 is necessary for the maintenance of GnRH neuron activity, at least in males, then factors responsible for a decline in GnRH mRNA might be the result of altered tet2 activity. BPA is an environmental contaminant shown to influence sperm quality and GnRH mRNA levels in adult male rats (33, 34). We evaluated the impact of BPA on tet2 subcellular localization and the GnRH gene chromatin environment in GT1–7 cells. After 2 hours of BPA exposure (10nM), tet2 exhibited a dramatic shift in subcellular localization (Figure 8A). Specifically, whereas tet2 immunoreactivity was largely distributed in both the nucleus and cytoplasm of vehicle treated cells; it dramatically shifted to primarily the cytoplasm during BPA exposure (Figure 8B). This shift was associated with modifications to the GnRH gene, particularly H3K4me3 abundance (Figure 8B). Tet2 binding trended toward a decrease in promoter region binding after 2 hours of BPA (10nM) exposure (two-way ANOVA; BPA vs vehicle exposure, F1,16 = 5.055, P = .039; gene region, F3,16 = 63.71, P < .001; interaction, F3,16 = 2.594, P = .0886) (Figure 8B). This was accompanied by a significant reduction in promoter H3K4me3 abundance (two-way ANOVA; BPA vs vehicle, F1,16 = 31.68, P < .001; gene region, F3,16 = 87.25, P < .001; interaction, F3,16 = 19.31, P < .001) (Figure 8B). There was no significant effect of this BPA exposure on H3K27me3 abundance at the regions evaluated. GnRH mRNA levels did not change during this brief 2-hour period of BPA exposure, although they were substantially lower after 12 hours of sustained BPA exposure (relative GnRH expression in GT1–7 cells by qPCR (2−ΔCT × 10−5) vehicle: 3.72 ± 0.09, BPA 10nM 2 h: 3.21 ± 0.19, BPA 10nM 12 h: 1.95 ± 0.14; n = 3 for each culture condition, ANOVA F2,6 = 39.03, P < .001 Tukey post hoc: vehicle = BPA 2 h, vehicle > BPA 12 h, BPA 2 h > BPA 12 h).

Figure 8.

Figure 8.

BPA exposure alters tet2 subcellular localization and the GnRH gene chromatin environment in GT1–7 cells. A, Immunofluorescence analysis of GT1–7 cells using tet2 antibody after BPA (10nM) or vehicle exposure for 2 hours. B, Quantification of tet2 immunoreactivity in 150 cells; immunoreactivity was categorized as occurring in the cytosol, the nucleus, or both compartments. C, Schematic diagram (adapted from Iyer et al [3]) of the mouse GnRH gene depicting locations of enhancers E3, E2, E1, and the GnRH promoter (P). Opposing arrows show ChIP assay primer binding locations. Fixed chromatin from GT1–7 cells exposed to either vehicle or BPA (10nM for 2 h) was sonicated then immunoprecipitated with antibodies against Tet2, H3K4me3, or H3K27me3. DNA was then analyzed by qPCR. Data are presented as fold enrichment relative to IgG (2 (IgG Ct-specific antibody Ct), mean of 3 cultures per cell line ± SEM. Regions not assigned the same letter are significantly different by Tukey post hoc; P < .05 after two-way ANOVA.

Tet2 protein interactions in GnRH neurons

The shift in tet2 subcellular localization during BPA exposure implicates the presence of a tet2 chaperone and/or cytoplasmic tet2 target in GnRH neurons. Although tet2 interactions have been reported for other cellular systems (23, 35, 36), there is presently no report of neuronal tet2-interacting proteins. Using mass spectrometry after coimmunoprecipitation, we identified novel tet2 interactions, which isolated the SET complex as likely responsible for H3K4me3 deposition at the GnRH promoter, as well as cytoplasmic targets potentially related to the BPA-driven shift in subcellular localization. The primary interactions, shown in Figure 9, suggest 4 potential roles of tet2 in GnRH neurons, including chromatin remodeling/histone modification, DNA repair, mitochondrial function, and RNA metabolism.

Figure 9.

Figure 9.

Tet2-interacting proteins in GT1–7 cells. Abundance of unique peptides identified by mass spectrometry from tet2 coimmunoprecipitates. The most prominent tet2 interactions are grouped into cellular functions associated with the interacting protein.

Discussion

These studies reveal tet2 as a key factor responsible for GnRH neuron activity and male reproductive function. Specifically, tet2 stimulates GnRH gene expression and peptide release from immature GnRH cells, and perhaps more importantly, maintains differentiated neuronal expression of GnRH mRNA and adult male reproductive function (Figure 10).

Figure 10.

Figure 10.

Tet2 influences accumulation and maintenance of the activating H3K4 trimethylation in immature and differentiated GnRH neurons, respectively. Top portion, Immature GnRH neuronal cell lines (GN11) express GnRH at very low levels and less tet2 than differentiated GnRH neuronal cell lines (GT1–7). Increasing tet2 expression in GN11 cells by transient transfection leads to an elevation of GnRH mRNA and H3K4me3 at the GnRH gene neuron-specific enhancer and promoter. Bottom portion, Knockdown of tet2 expression in differentiated GnRH neuronal cultures (GT1–7 cells) leads to a loss of tet2 binding near the GnRH gene promoter, loss of H3K4me3 abundance in the same region, and reduction in GnRH mRNA levels.

Despite our initial hypotheses and in vitro findings in immature GN11 cells, we did not find an effect of specific tet2 ablation in GnRH neurons on pubertal timing. However, the apparent discrepancies between in vitro and in vivo results may be explained by limitations of the Cre-lox approach as applied to GnRH neuronal gene targeting in these studies. Specifically, the Cre-mediated excision and consequential inactivation of tet2 activity likely occurs after the embryonic period of GnRH gene activation. The Cre transgene in these mice is driven by a fragment of the mouse GnRH promoter (−3446/+28); this spans the regulatory region analogous to the primate gene segment that undergoes developmental changes in DNA methylation status (2) as well as the regions bound by tet2 and exhibiting developmental changes in H3K4me3 abundance. If in fact this region were targeted by tet2 in GnRH neurons during development, then tet2-mediated reorganization would simultaneously occur at the transgene promoter and the endogenous GnRH promoter. Consequently, endogenous embryonic GnRH promoter reorganization would be complete before ablation of tet2 activity. Because gTKO animals showed no evidence of pubertal delay, we speculate that postnatal tet2 activity is not necessary for a pubertal rise in GnRH neurosecretion. Whether another tet enzyme compensates for lost tet2 activity in this context remains unknown.

Mellon and coworkers (3) previously discovered that the GnRH gene promoter and enhancer regions in immature GN cells were more heavily associated with a repressive histone modification: H3 lysine 9 (K9) di-methylation, whereas the same genomic regions in GT1 cells were associated with the permissive H3K9 acetylation and H3K4me3 modifications. Importantly, treatment of GN cells with a histone deacetylase inhibitor had no impact on gene expression, suggesting a mechanism other than acetylation of histones must be responsible for activation of GnRH transcription. Our data suggest that tet2 contributes to the establishment of a permissive chromatin state. Further, based on our findings in GT1–7 cells with disrupted tet2 expression and gTKO animals, tet2 appears to factor most heavily in the maintenance of a postpubertal permissive chromatin structure and adult male reproductive function.

Because of the apparent restriction of reproductive dysfunction to male gTKO animals, we speculate that an elevation of plasma testosterone (T) may be responsible for driving postpubertal GnRH gene chromatin reorganization towards a less permissive state. Importantly, although T already has a suppressive effect on LH release before puberty, GnRH mRNA levels are not sensitive to the suppressive impact of T until after puberty (37). In addition, T signals, at least in part, through protein kinase C to influence GnRH release (38). Importantly, protein kinase C activation closes chromatin in GT1 cells, causing a reduction in GnRH promoter H3 acetylation, accumulation of H3K9 methylation and suppression of GnRH gene expression (3). In order to maintain GnRH gene expression levels, there would need to be a reversal of T-mediated chromatin closure. Based on our present data, we suspect that tet2-mediated deposition of H3K4me3 could overcome a T-mediated transient shift toward a less permissive chromatin environment. In that case, however, gTKO animals would be incapable of reversing the strengthening T-mediated restriction of chromatin near the GnRH gene after puberty. Consequently, these animals would suffer the consequences observed in our aging gTKO males, that is, reduced hypothalamic GnRH mRNA, declining plasma LH, and less efficient reproductive function.

These studies focused on tet2 activity in GnRH neurons based on our initial discoveries that tet2 expression 1) was significantly lower in GN11 compared with GT1–7 cells, and 2) increased during prepubertal development in the hypothalamus of male and female mice as well as the female rhesus monkey (J. R. Kurian and E. Terasawa, unpublished observation). Critical questions remain as to how tet2 expression itself is regulated. Recent evidence suggests that tet2 expression is under control of a CpG-rich regulatory element (39). Methylated CpG islands recruit polycomb group (PcG) and zinc finger (ZNF) proteins to restrict associated gene expression (4042). Importantly, Lomniczi et al have recently provided evidence that both PcG (43) and ZNF protein (44) complexes have substantial influence over the neuroendocrine control of puberty onset and reproductive function. Whether PcG or ZNF complexes alter tet2 expression in the hypothalamus is not reported; however, the Zinc-finger CXXC domain containing protein IDAX is responsible for down-regulation of tet2 in HEK293T cells (45). All together, these findings may implicate a complex and intriguing interplay between tet2, PcG, and ZNF proteins in the hypothalamic control of reproduction.

We show that removing tet2 from GT1–7 cells led to a less permissive chromatin environment at the GnRH gene and a decline in GnRH mRNA. Furthermore, losing tet2 activity in GnRH neurons caused a drop in adult male hypothalamic GnRH mRNA content, plasma LH, and reproductive performance. Together, these findings suggest that tet2 is necessary for the maintenance of elevated GnRH mRNA levels and neuronal activity in general. Important questions now surface; that is, under what conditions is tet2 activity compromised and how might this relate to a decline in reproductive function? We consider these questions in the context of 2 factors known to negatively impact hypothalamic levels of GnRH mRNA and reproductive function, namely exposure to environmental contaminants and aging.

BPA is a pervasive environmental contaminant, already detectable in 93% of Americans (46) with world-wide production increasing annually (47). Although currently considered a potential hazard during development, evidence is accumulating for direct effects of BPA exposure on adult physiology. In terms of reproduction, male adult rats exposed to BPA are reported to have abnormal plasma LH levels, reduced sperm quality (34) and decreased hypothalamic GnRH mRNA content (33). We exposed GT1–7 cells to BPA at a concentration presumed relevant to high-level adult human exposure (48, 49). The impact on tet2 was profound, shifting localization primarily to the cytoplasm. This shift was accompanied by a loss of H3K4me3 abundance at the GnRH gene promoter. That loss of H3K4me3 is consistent with the findings of Mellon and coworkers (3), who reported a loss of permissive chromatin status in the GnRH gene 5′ region after exposure of GT1–7 cells to 12-O-tetradecanoylphorbol-13-acetate. Importantly, BPA and 12-O-tetradecanoylphorbol-13-acetate are both potent generators of superoxide (50, 51) and mitochondrial stress. Given our current findings of significant tet2 association with mitochondrial enzymes, we suggest that BPA generated oxidative stress could initiate recruitment of tet2 to mitochondria. This mitochondrial recruitment in response to oxidative stress would be a novel tet2 mechanism of action, although also consistent with the previously reported presence of hydroxymethylated mitochondrial DNA (52) and role of tet1 in fighting oxidant induced neuronal cell death (53). A pertinent deleterious consequence of this mitochondrial recruitment would be decreased association of tet2 with nuclear genes (eg, GnRH) and consequential loss of a permissive chromatin environment at those genes.

The potential role of tet2 in response to oxidative stress is also consistent with the necessity of tet2 for abatement of inflammatory processes (36). In acting as a terminator of inflammatory processes, tet2 could have direct implications on GnRH neuronal activity in the aging hypothalamus. A recent report suggests that elevated hypothalamic inflammation with increasing age is responsible for declining GnRH neuron activity, leading to age related cognitive decline and physical capabilities. In those studies, a model of constitutive hypothalamic inflammation enabled constant activation of NFκB in hypothalamic neurons, leading to a decline in GnRH mRNA levels and premature systemic aging (6). Notably, Tet2 was recently found necessary for termination of NFκB-initiated inflammatory gene expression in bone marrow-derived stem cells (36). This Tet2-mediated termination of expression required gene repression through association with a histone deacetylase enzyme (Hdac). Our current protein association data indicates an interaction between hdac1 and tet2 in GT1–7 cells, suggesting that tet2 could also influence the repression of gene expression across portions of the GnRH neuron genome. T, this suggests that under conditions of elevated hypothalamic inflammation such as aging or obesity, tet2 could be recruited to genomic regions targeted for repression and resolution of neuronal inflammation, thereby vacating the GnRH gene, again with consequential loss of permissive chromatin modifications in that region.

The present interactome data, to our knowledge, are the first reported tet2 interactions within a neuronal cell. These data provide additional evidence for recently described tet2 activities but also suggest novel roles for tet enzymes in neuronal function. Tet2 has already been shown to promote H3K4me3 through the SET1/COMPASS complex (23). Consequently, we were not surprised to find tet2 interactions with wdr82 and wdr5, 2 major components of the mammalian SET1 complex. Importantly, the inclusion of wdr82, which is not found in the menin/MLL complex, seems to isolate SET1 as the tet2 associated H3K4 methylating complex in GnRH cells.

Although not previously reported, interactions with some DNA repair enzymes align with the major presumed role of tet2; that being active DNA demethylation, which was the initial motivation to investigate tet activity in GnRH neurons. Active DNA demethylation is commonly thought to require initiation through oxidation of methyl-cytosine by tet enzymes, followed by recognition/DNA excision and subsequent repair. Consequently, interaction with the mismatch repair protein msh6, and the DNA double-strand break-binding protein Ddb1, is predictable.

On the other hand, the associations between tet2 and enzymes responsible for RNA metabolism, particularly RNA splicing, were unexpected although potentially integral to GnRH neuron activity. Recent evidence indicates tet2-mediated intragenic DNA hydroxymethylation is critical to RNA transcript splicing through CTCF (54). Importantly, mature GnRH mRNA requires excision of the first intron. Excision of this intron from GnRH pre-mRNA is developmentally distinct (55, 56), with generation of the mature mRNA most prominent in adult rodents. In fact, although mature GnRH mRNA levels are higher in adult compared with prepubertal female rats, this appears to be the consequence of posttranscriptional processing, as GnRH pre-mRNA levels do not differ between those developmental stages (57). In addition, splicing of the first intron from GnRH pre-mRNA is largely attenuated in hypogonadal mice (58), suggesting conditions that alter splicing efficiency could have a substantial impact on reproductive function. Whether tet2 activity or DNA hydroxymethylation are related to GnRH pre-mRNA splicing efficiency and consequently reproductive function remains an open question, although the association of tet2 with splicing factors in GT1–7 cells raises that intriguing possibly.

Conclusions

In these studies we show that tet2 activity in GnRH neurons has influence over the neuroendocrine control of male reproductive function. We provide evidence that tet2 activity may not be restricted to epigenome manipulation. Specifically, tet2 may participate in mitochondrial function, RNA splicing, and gene repression. Importantly, these novel neuronal tet2 activities may be protective responses to environmental conditions including aging, obesity, and exposure to environmental contaminants. Taken as a whole, these studies isolate an enzyme responsible for epigenetic regulation of the neuronal GnRH gene; they also provide novel insight toward the relationship between the environment, GnRH neuron activity and related physiology.

Acknowledgments

GnRH cell lines were provided by Dr Pamela Mellon (GT1–7; University of California, San Diego) and Dr Sally Radovick (GN11; Rutgers University). We thank Dr Hemanta Shrestha for preparation of iodinated peptide, Dr Nicholas Sanek and Dr Sharon Dubois for assistance with tissue collections, and Ms Jessica Felton for assistance with animal husbandry and genotyping.

This work was supported by Public Health Service grants from the National Institutes of Health R00ES020878 (to J.R.K.), 2R01HD011355 (to E.T.), and 1R01HD068777 (to J.E.L.). This work was also supported by the National Science Foundation IOS Grant 1049849 “Research Coordination Network for Epigenetics, Behavior and Reproduction.”

Disclosure Summary: The authors have nothing to disclose.

For News & Views see page 3398

Abbreviations:
BPA
bisphenol A
ChIP
chromatin-immunoprecipitation
CON
control
Ct
critical threshold
gTKO
GnRH-specific tet2 knockout
H3K4me3
histone 3 lysine 4 trimethylation
5hmC
5-hydroxymethylcytosine
K9
lysine 9
KO
knockout
p
postnatal day
PcG
polycomb group
POA-MBH
preoptic area and mediobasal hypothalamic
T
testosterone
tet
ten-eleven translocation
V.O.
vaginal opening
ZNF
zinc finger.

References

  • 1. Kurian JR, Keen KL, Terasawa E. Epigenetic changes coincide with in vitro primate GnRH neuronal maturation. Endocrinology. 2010;151:5359–5368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Kurian JR, Terasawa E. Epigenetic control of gonadotropin releasing hormone neurons. Front Endocrinol (Lausanne). 2013;4:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Iyer AK, Brayman MJ, Mellon PL. Dynamic chromatin modifications control GnRH gene expression during neuronal differentiation and protein kinase C signal transduction. Mol Endocrinol. 2011;25:460–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Novaira HJ, Yates M, Diaczok D, Kim H, Wolfe A, Radovick S. The gonadotropin-releasing hormone cell-specific element is required for normal puberty and estrous cyclicity. J Neurosci. 2011;31:3336–3343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Kermath BA, Riha PD, Woller MJ, Wolfe A, Gore AC. Hypothalamic molecular changes underlying natural reproductive senescence in the female rat. Endocrinology. 2014;155:3597–3609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Zhang G, Li J, Purkayastha S, et al. Hypothalamic programming of systemic ageing involving IKK-β, NF-κB and GnRH. Nature. 2013;497:211–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Tahiliani M, Koh KP, Shen Y, et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science. 2009;324:930–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Ito S, D'Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010;466:1129–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Ito S, Shen L, Dai Q, et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science. 2011;333:1300–1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Maiti A, Drohat AC. Thymine DNA glycosylase can rapidly excise 5-formylcytosine and 5-carboxylcytosine: potential implications for active demethylation of CpG sites. J Biol Chem. 2011;286:35334–35338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. He YF, Li BZ, Li Z, et al. Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science. 2011;333:1303–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Wu H, D'Alessio AC, Ito S, et al. Genome-wide analysis of 5-hydroxymethylcytosine distribution reveals its dual function in transcriptional regulation in mouse embryonic stem cells. Genes Dev. 2011;25:679–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Williams K, Christensen J, Pedersen MT, et al. TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature. 2011;473:343–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Pastor WA, Pape UJ, Huang Y, et al. Genome-wide mapping of 5-hydroxymethylcytosine in embryonic stem cells. Nature. 2011;473:394–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Ficz G, Branco MR, Seisenberger S, et al. Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature. 2011;473:398–402. [DOI] [PubMed] [Google Scholar]
  • 16. Xu Y, Wu F, Tan L, et al. Genome-wide regulation of 5hmC, 5mC, and gene expression by Tet1 hydroxylase in mouse embryonic stem cells. Mol Cell. 2011;42:451–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Stroud H, Feng S, Morey Kinney S, Pradhan S, Jacobsen SE. 5-Hydroxymethylcytosine is associated with enhancers and gene bodies in human embryonic stem cells. Genome Biol. 2011;12:R54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Yu M, Hon GC, Szulwach KE, et al. Base-resolution analysis of 5-hydroxymethylcytosine in the mammalian genome. Cell. 2012;149:1368–1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Sun Z, Terragni J, Jolyon T, et al. High-resolution enzymatic mapping of genomic 5-hydroxymethylcytosine in mouse embryonic stem cells. Cell Rep. 2013;3:567–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Yildirim O, Li R, Hung JH, et al. Mbd3/NURD complex regulates expression of 5-hydroxymethylcytosine marked genes in embryonic stem cells. Cell. 2011;147:1498–1510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Mellén M, Ayata P, Dewell S, Kriaucionis S, Heintz N. MeCP2 binds to 5hmC enriched within active genes and accessible chromatin in the nervous system. Cell. 2012;151:1417–1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Spruijt CG, Gnerlich F, Smits AH, et al. Dynamic readers for 5-(hydroxy)methylcytosine and its oxidized derivatives. Cell. 2013;152:1146–1159. [DOI] [PubMed] [Google Scholar]
  • 23. Deplus R, Delatte B, Schwinn MK, et al. TET2 and TET3 regulate GlcNAcylation and H3K4 methylation through OGT and SET1/COMPASS. EMBO J. 2013;32:645–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Wong ML, Medrano JF. Real-time PCR for mRNA quantitation. BioTechniques. 2005;391:75–85. [DOI] [PubMed] [Google Scholar]
  • 25. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3:1101–1108. [DOI] [PubMed] [Google Scholar]
  • 26. Moran-Crusio K, Reavie L, Shih A, et al. Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell. 2011;20(1):11–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Wolfe A, Divall S, Singh SP, et al. Temporal and spatial regulation of CRE recombinase expression in gonadotrophin-releasing hormone neurones in the mouse. J Neuroendocrinol. 2008;20:909–916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Terasawa E, Keen KL, Mogi K, Claude P. Pulsatile release of luteinizing hormone-releasing hormone in cultured LHRH neurons derived from the embryonic olfactory placode of the rhesus monkey. Endocrinology. 1999;140:1432–1441. [DOI] [PubMed] [Google Scholar]
  • 29. Martínez de la Escalera G, Choi AL, Weiner RI. Generation and synchronization of gonadotropin-releasing hormone (GnRH) pulses: intrinsic properties of the GT1–1 GnRH neuronal cell line. Proc Natl Acad Sci USA. 1992;89:1852–1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Gearing M, Terasawa E. Luteinizing hormone releasing hormone (LHRH) neuroterminals mapped using the push-pull perfusion method in the rhesus monkey. Brain Res Bull. 1988;21:117–121. [DOI] [PubMed] [Google Scholar]
  • 31. Radovick S, Wray S, Lee E, et al. Migratory arrest of gonadotropin-releasing hormone neurons in transgenic mice. Proc Natl Acad Sci USA. 1991;88:3402–3406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Mellon PL, Windle JJ, Goldsmith PC, Padula CA, Roberts JL, Weiner RI. Immortalization of hypothalamic GnRH neurons by genetically targeted tumorigenesis. Neuron. 1990;5:1–10. [DOI] [PubMed] [Google Scholar]
  • 33. Jin P, Wang X, Chang F, et al. Low dose bisphenol A impairs spermatogenesis by suppressing reproductive hormone production and promoting germ cell apoptosis in adult rats. J Biomed Res. 2013;27:135–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Wisniewski P, Romano RM, Kizys MM, et al. Adult exposure to bisphenol A (BPA) in Wistar rats reduces sperm quality with disruption of the hypothalamic-pituitary-testicular axis. Toxicology. 2015;329:1–9. [DOI] [PubMed] [Google Scholar]
  • 35. Wang Y, Xiao M, Chen X, et al. WT1 recruits TET2 to regulate its target gene expression and suppress leukemia cell proliferation. Mol Cell. 2015;57:662–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Zhang Q, Zhao K, Shen Q, et al. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature. 2015;525:389–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Richardson HN, Gore AC, Venier J, Romeo RD, Sisk CL. Increased expression of forebrain GnRH mRNA and changes in testosterone negative feedback following pubertal maturation. Mol Cell Endocrinol. 2004;214:63–70. [DOI] [PubMed] [Google Scholar]
  • 38. Wetsel WC, Negro-Vilar A. Testosterone selectively influences protein kinase-C-coupled secretion of proluteinizing hormone-releasing hormone-derived peptides. Endocrinology. 1989;125:538–547. [DOI] [PubMed] [Google Scholar]
  • 39. Sohni A, Bartoccetti M, Khoueiry R, et al. Dynamic switching of active promoter and enhancer domains regulates Tet1 and Tet2 expression during cell state transitions between pluripotency and differentiation. Mol Cell Biol. 2015;35:1026–1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Riising EM, Comet I, Leblanc B, Wu X, Johansen JV, Helin K. Gene silencing triggers polycomb repressive complex 2 recruitment to CpG islands genome wide. Mol Cell. 2014;55:347–360. [DOI] [PubMed] [Google Scholar]
  • 41. Choo Y. Recognition of DNA methylation by zinc fingers. Nat Struct Biol. 1998;5:264–265. [DOI] [PubMed] [Google Scholar]
  • 42. Liu Y, Zhang X, Blumenthal RM, Cheng X. A common mode of recognition for methylated CpG. Trends Biochem Sci. 2013;38:177–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Lomniczi A, Loche A, Castellano JM, et al. Epigenetic control of female puberty. Nat Neurosci. 2013;16:281–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Lomniczi A, Wright H, Castellano JM, et al. Epigenetic regulation of puberty via Zinc finger protein-mediated transcriptional repression. Nat Commun. 2015;6:10195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Ko M, An J, Bandukwala HS, et al. Modulation of TET2 expression and 5-methylcytosine oxidation by the CXXC domain protein IDAX. Nature. 2013;497:122–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Calafat AM, Ye X, Wong LY, Reidy JA, Needham LL. Exposure of the U.S. population to bisphenol A and 4-tertiary-octylphenol: 2003–2004. Environ Health Perspect. 2008;116:39–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Global Industry Analysts, Inc. 2010 bisphenol A: a global strategic business report. Available from http://www.strategyr.com/Bisphenol_A_Market_Report.asp Accessed July 2015.
  • 48. Vandenberg LN, Chahoud I, Heindel JJ, Padmanabhan V, Paumgartten FJ, Schoenfelder G. Urinary, circulating, and tissue biomonitoring studies indicate widespread exposure to bisphenol A. Environ Health Perspect. 2010;118:1055–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Kurian JR, Keen KL, Kenealy BP, Garcia JP, Hedman CJ, Terasawa E. Acute influences of bisphenol A exposure on hypothalamic release of gonadotropin-releasing hormone and kisspeptin in female rhesus monkeys. Endocrinology. 2015;156:2563–2570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Swindle EJ, Hunt JA, Coleman JW. A comparison of reactive oxygen species generation by rat peritoneal macrophages and mast cells using the highly sensitive real-time chemiluminescent probe pholasin: inhibition of antigen-induced mast cell degranulation by macrophage-derived hydrogen peroxide. J Immunol. 2002;169:5866–5873. [DOI] [PubMed] [Google Scholar]
  • 51. Huang R, Zhao L, Chen H, et al. Megakaryocytic differentiation of K562 cells induced by PMA reduced the activity of respiratory chain complex IV. PLoS One. 2014;9:e96246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Shock LS, Thakkar PV, Peterson EJ, Moran RG, Taylor SM. DNA methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mammalian mitochondria. Proc Natl Acad Sci USA. 2011;108:3630–3635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Xin Y, Yuan B, Yu B, et al. Tet1-mediated DNA demethylation regulates neuronal cell death induced by oxidative stress. Sci Rep. 2015;5:7645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Marina RJ, Sturgill D, Bailly MA, et al. TET-catalyzed oxidation of intragenic 5-methylcytosine regulates CTCF-dependent alternative splicing. EMBO J. 2015;e201593235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Seong JY, Park S, Kim K. Enhanced splicing of the first intron from the gonadotropin-releasing hormone (GnRH) primary transcript is a prerequisite for mature GnRH messenger RNA: presence of GnRH neuron-specific splicing factors. Mol Endocrinol. 1999;13:1882–1895. [DOI] [PubMed] [Google Scholar]
  • 56. Son GH, Jung H, Seong JY, Choe Y, Geum D, Kim K. Excision of the first intron from the gonadotropin-releasing hormone (GnRH) transcript serves as a key regulatory step for GnRH biosynthesis. J Biol Chem. 2003;278:18037–18044. [DOI] [PubMed] [Google Scholar]
  • 57. Gore AC, Wu TJ, Rosenberg JJ, Roberts JL. Gonadotropin-releasing hormone and NMDA receptor gene expression and colocalization change during puberty in female rats. J Neurosci. 1996;16:5281–5289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Seong JY, Kim BW, Park S, Son GH, Kim K. First intron excision of GnRH pre-mRNA during postnatal development of normal mice and adult hypogonadal mice. Endocrinology. 2001;142:4454–4461. [DOI] [PubMed] [Google Scholar]

Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES