Open Access
How to translate text using browser tools
19 January 2011 GATA4 Deficiency Impairs Ovarian Function in Adult Mice
Antti Kyrönlahti, Melanie Vetter, Rosemarie Euler, Malgorzata Bielinska, Patrick Y. Jay, Mikko Anttonen, Markku Heikinheimo, David B. Wilson
Author Affiliations +
Abstract

Transcription factor GATA4 is expressed in granulosa cells and, to a lesser extent, in other ovarian cell types. Studies of mutant mice have shown that interactions between GATA4 and its cofactor, ZFPM2 (also termed FOG2), are required for proper development of the fetal ovary. The role of GATA4 in postnatal ovarian function, however, has remained unclear, in part because of prenatal lethality of homozygous mutations in the Gata4 gene in mice. To circumvent this limitation, we studied ovarian function in two genetically engineered mouse lines: C57BL/6 (B6) female mice heterozygous for a Gata4-null allele, and 129;B6 female mice in which Gata4 is deleted specifically in proliferating granulosa cells using the Cre-loxP recombination system and Amhr2-cre. Female B6 Gata4 /− mice had delayed puberty but normal estrous cycle lengths and litter size. Compared to wild-type mice, the ovaries of gonadotropin-stimulated B6 Gata4 /− mice were significantly smaller, released fewer oocytes, produced less estrogen, and expressed less mRNA for the putative GATA4 target genes Star, Cyp11a1, and Cyp19. Gata4 conditional knockout (cKO) mice had a more severe phenotype, including impaired fertility and cystic ovarian changes. Like Gata4 /− mice, the ovaries of gonadotropin-stimulated cKO mice released fewer oocytes and expressed less Cyp19 than those of control mice. Our findings, coupled with those of other investigators, support the premise that GATA4 is a key transcriptional regulator of ovarian somatic cell function in both fetal and adult mice.

INTRODUCTION

Transcription factor GATA4 is implicated in the development and function of the mammalian ovary [13]. GATA4 is expressed in pregranulosa and stroma cells of the fetal ovary and in granulosa and theca cells of the postnatal ovary [46]. In adult mice, GATA4 is thought to play a role in the maintenance or maturation of granulosa cells; Gata4 is expressed in potentially mitotic and proliferating granulosa cells but is downregulated when proliferation ceases during ovulation, apoptosis, or luteinization [4, 7]. Follicle-stimulating hormone (FSH), the gonadotropin that initiates follicular growth, increases both the expression level and the intrinsic activity of GATA4 in granulosa cells [4, 8, 9]. Conversely, loss-of-function mutations in the FSH receptor are associated with reduced ovarian expression of GATA4 in both mice [10] and humans [11]. GATA4 binds and activates genes essential for ovarian steroidogenesis, including Star [12, 13], Cyp11a1 [14], and Cyp19 (aromatase) [9, 1518].

Analysis of genetically engineered mice has shown that interactions between GATA4 and its cofactor, ZFPM2 (also termed FOG2), are necessary for early ovarian development [3, 19]. Zfpm2−/− mice and Gata4ki/ki mice, which bear a knockin mutation that abrogates the interaction of GATA4 with ZFPM2, exhibit identical fetal ovarian phenotypes [3, 19] that include decreased expression of genes essential for early ovarian development, such as Wnt4 [20] and Foxl2 [21]. The ovaries of Zfpm2−/− and Gata4ki/ki fetal mice also express excess DKK1, a secreted inhibitor of the canonical WNT/β-catenin pathway, a signaling cascade critical for female gonadogenesis [3, 19].

Although GATA4 appears to be essential for proper fetal ovarian development, the role of this transcription factor in postnatal ovarian function remains unclear, in part because of prenatal lethality of homozygous mutations in the Gata4 gene in mice [2227]. Adult transgenic mice expressing a tetracycline-inducible small interfering RNA (siRNA) directed against GATA4 appear to have intact ovarian steroidogenesis, insofar as circulating levels of FSH and luteinizing hormone (LH) are normal [28]. Approximately 10% of GATA4 siRNA transgenic mice develop ovarian teratomas, suggesting that GATA4 functions as a tumor suppressor in this tissue [28].

In the present study, we examine the impact of GATA4 deficiency on ovarian physiology using two complementary mouse models: C57BL/6 (B6) female mice heterozygous for a germline deletion mutation in Gata4, and 129;B6 female mice in which Gata4 is conditionally ablated in proliferating granulosa cells using the Cre-loxP recombination system.

MATERIALS AND METHODS

Experimental Mice

Procedures involving mice were approved by the Washington University Institutional Committee for Laboratory Animal Care and were conducted in accordance with the National Research Council publication Guide for Care and Use of Laboratory Animals. All mice had free access to water and standard rodent chow and were exposed to 12L:12D photoperiods. Gata4+/ mice, originally produced by William Pu, were genotyped as described previously [29, 30]. These mice were backcrossed with B6 mice (Charles River Laboratories) for a minimum of 10 generations. Gata4Flox/Flox mice (also termed Gata4tm1.1Sad/J), originally generated by Stephen Duncan [27, 31], were purchased from the Jackson Laboratory and genotyped as described previously [27, 31]. The 129;B6 Amhr2cre/+ mice (also termed B6;129S7-Amhr2tm3(cre)Bhr/Mmnc), originally produced by Richard Behringer, were obtained from the Mutant Mouse Regional Resource Centers (MMRRC) and genotyped as described previously [32, 33]. ROSA26 Flox-stop-Flox LacZ reporter mice (also termed B6.129S4-Gt(ROSA)26Sortm1Sor/J), developed by P. Soriano [34], were obtained from the Jackson Laboratory. To generate conditional knockout (cKO) mice, Gata4Flox/Flox mice were mated with Amhr2cre/+ mice, and the resultant Gata4Flox/+;Amhr2cre/+ mice were mated with Gata4Flox/Flox mice to produce Gata4Flox/Flox;Amhr2cre/+ mice.

Measurement of Pubertal Events

Daily vaginal lavage was begun on the day of vaginal opening and continued for the duration of the present study [35]. Cytologic analysis was performed on air-dried, Giemsa-stained smears. Estrous cycle stage was determined by the presence of nucleated epithelial cells (NEC), cornified epithelial cells (CEC), and polymorphonuclear cells (PMN), with estrus characterized by CEC and some NEC without PMN, metestrus by CEC and PMN, diestrus by PMN with few NEC, and proestrus mainly by NEC and few CEC and PMN. Ambiguous cycle status was ascertained based on determinations for the preceding and subsequent day and were labeled as an intermediate value. The age of first vaginal cornification and the onset of estrous cyclicity were determined as described previously [35]. Cycle length was defined as the time between onsets of estrus.

Fertility Studies

Reproductive performance was assessed by housing female mice with proven male stud mice and measuring litter size over successive pregnancies. Because some Gata4+/− mice die within days of birth because of developmental defects in the heart, lungs, or diaphragm [3638], we assessed litter size on the morning of birth.

Tissue Isolation and Histological Analyses

At specified times, mice were killed by carbon dioxide inhalation. Blood was collect by cardiac puncture. Ovaries or uteri were harvested, fixed overnight in 4% paraformaldehyde in PBS, and embedded in paraffin for routine histology. Paraffin sections (thickness, 5 μm) were stained with hematoxylin-and-eosin or subjected to immunoperoxidase staining with goat anti-mouse GATA4 immunoglobulin (Ig) G (sc-1237; Santa Cruz Biotechnology, Inc.) at a 1:200 dilution. The secondary antibody employed for immunoperoxidase staining was donkey anti-goat biotinylated IgG (Jackson ImmunoResearch) at a 1:1000 dilution. The avidin-biotin immunoperoxidase system (Vectastain Elite ABC Kit; Vector Laboratories, Inc.) and diaminobenzidine (Sigma-Aldrich Corp.) were used to visualize the bound antibody; slides were then counterstained with toluidine blue. For X-gal staining, frozen tissue sections (thickness, 10 μm) were prepared after embedding mouse ovaries in O.C.T. (Tissue-Tek). Sections were fixed with 0.2% glutaraldehyde for 10 min; permeabilized with 100 mM potassium phosphate (pH 7.4), 0.02% NP-40, and 0.01% sodium deoxycholate for 5 min; and then incubated in 0.5 mg/ml of X-gal (Sigma-Aldrich Corp.) with 10 mM K3[Fe(CN)6], 10 mM K4[Fe(CN)6], 100 mM potassium phosphate (pH 7.4), 0.02% NP-40, and 0.01% sodium deoxycholate at 37°C overnight [25]. X-gal-stained sections were counterstained with eosin.

Ovarian Follicle Counts

Oocyte-containing follicles were scored as described previously [39, 40]. Briefly, ovaries were collected from mice shortly after the onset of puberty, fixed (0.34 N glacial acetic acid, 28% ethanol, and 10% formalin), embedded in paraffin, serially sectioned (thickness, 6 μm), and stained with hematoxylin-picric acid methyl blue. The number of ostensibly healthy and atretic primordial, primary, and small preantral follicles was determined in every fifth section through the entire ovary. Primordial follicles contained a compact oocyte enveloped by a single layer of flattened granulosa cells. Primary follicles had an enlarged oocyte surrounded by a single layer of cuboidal granulosa cells. Small preantral follicles had an enlarged oocyte surrounded by two to four layers of cuboidal granulosa cells but no visible antrum. Only those follicles in which the oocyte nucleus was visible were scored, and follicles were designated atretic if the oocyte was degenerated, using criteria delineated elsewhere [39, 40]. The total number of healthy and atretic follicles per ovary was then calculated as described previously [39, 40].

Gonadotropin and Estradiol Treatment

Immature (age, 19–25 days) mice were induced to ovulate using equine chorionic gonadotropin (eCG; 5 IU i.p.), followed by human chorionic gonadotropin (hCG; 5 IU i.p.) 48 h later. Four treatment groups were analyzed: 1) no stimulation; 2) eCG for 48 h (follicular growth); 3) eCG for 48 h, followed by hCG for 18 h (postovulation); and 4) eCG for 48 h, followed by hCG for 5 days (luteal glands). Harvested ovaries were processed for histological analysis or RNA isolation. To quantify ovulation, mice were killed 18 h after hCG administration, and oocytes were isolated from oviducts [41].

RNA Isolation and First-Strand cDNA Synthesis

Total RNA was isolated from ovaries using TRIzol (Invitrogen). First-strand cDNA was produced with the SuperScript VILO cDNA Synthesis Kit (Invitrogen) using 700 ng of RNA as a template. An aliquot (1.5 μl) of cDNA was subjected to real-time RT-PCR using SYBR GreenER qPCR SuperMix (Invitrogen). Conditions for all quantitative RT-PCR (qRT-PCR) reactions were optimized in a light cycler (Stratagene Mx3005), which was programmed as follows: denaturation at 95°C for 10 min, followed by 40 cycles of amplification and quantification at 95°C for 30 sec and 60°C for 1 min with single fluorescent measurement, followed by melting at 60–95°C with a heating rate of 0.1°C/sec and continuous fluorescence measurement, followed by a final cooling step to 55°C. Melting curves did not reveal any significant contamination. The relative expression of target genes was calculated using the relative standard curve method as described in Guide to Performing Relative Quantitation of Gene Expression Using Real-Time Quantitative PCR (Applied Biosystems) and in our prior publication [42].

To compensate for variation among runs, the target gene expression was normalized to the expression of ribosomal protein L19. Primer pairs used for qRT-PCR assays were as follows: L19, NM_005084, (forward) 5′-GAAATCGCCAATGCCAACTC-3′, (reverse) 5′-TCTTAGACCTGCGAGCCTCA-3′, 405 bp; Cyp19, NM_007810, (forward) 5′-ATGTTCTTGGAAATGCTGAACCC-3′, (reverse) 5′-AGGACCTGGTATTGAAGACGAG-3′, 150 bp; Star, NM_011485, (forward) 5′-GCAGCAGGCAACCTGGTG-3′, (reverse) 5′-TGATTGTCTTCGGCAGCC-3′, 246 bp; Cyp11a1, NM_19779.3, (forward) 5′-CGATACTCTTCTCATGCGAG-3′, (reverse) 5′-CTTTCTTCCAGGCATCTGAAC-3′, 125 bp; Gata4, NM_008092.3, (for ward) 5′-CCCTACCCAGCCTACATGG-3′, (reverse) 5′-ACATATCGAGATTGGGGTGTCT-3′, 138 bp.

Semiquantitative RT-PCR for Detection of Cre-Mediated Recombination

First-strand cDNA was subjected to RT-PCR using a forward primer from exon 2 of the Gata4 gene, 5′-CCCTACCCAGCCTACATGG-3′, and a reverse primer from exon 7 of the same gene, 5′-GAGCTGGCCTGCGATGTCTGAGTG-3′, and the following reaction conditions: denaturation at 95°C for 10 min; followed by 35 cycles of amplification and quantification at 95°C for 30 sec, 55°C for 1 min, and 72°C for 1 min; followed by a final cooling step to 55°C. The intact Gata4Flox allele gave rise to a 782-bp band, whereas the recombined allele yielded a 401-bp band, reflecting deletion of exons 3–5.

Hormone Measurements

Serum estradiol (E2) levels were determined using a commercial radioimmunoassay (BioCheck). Serum antimüllerian hormone (AMH) levels were measured by ELISA as described elsewhere [43].

In Situ Hybridization

Tissue cryosections (thickness, 10 μm) were processed and subjected to mRNA in situ hybridization as described previously [25]. The sections were incubated with 1.2 × 106 cpm [33P]αUTP-labeled (1000–3000 Ci/mmol; Amersham) antisense riboprobe in a total volume of 80 μl. The riboprobe for mouse GATA4 was prepared as described previously [25].

Statistical Analyses

The effects of genotype on the age of first vaginal cornification, onset of estrous cyclicity, estrous cycle length, litter size, ovarian weight, uterine weight, steroid hormone levels, and prevalence of infertility were determined by unpaired, two-tailed t-tests. Statistical significance was set at P < 0.05. Two-way ANOVA was used to analyze the proportion of time in each estrous stage, follicle counts, and gene expression levels. Two-population-proportion testing was used to analyze the incidence of cystic ovarian changes. All numerical data are presented as the mean ± SD.

RESULTS

Delayed Puberty but Normal Estrous Cycle Length and Litter Size in Gata4+/− Female Mice

We first analyzed B6 mice heterozygous for a deletion in exon 2 of the Gata4 gene [29]. This germline mutation removes the translation start site and N-terminal activation domain of GATA4. Studies have shown that mice heterozygous for this allele, hereafter referred to as Gata4+/− mice, express wild-type (WT) Gata4 mRNA at half-normal levels in heart and other tissues in which this transcription factor is expressed [29, 36].

Vaginal cytology specimens were analyzed to determine the impact of Gata4 haploinsufficiency on the timing of two pubertal events: the first vaginal cornification and the onset of estrous cyclicity [44]. Previous studies have shown that vaginal cornification is dependent on an increase in circulating E2, whereas the onset of estrous cyclicity is dependent on both a preovulatory increase in plasma E2 and the ability of the pituitary to respond with a preovulatory surge of LH, which stimulates ovulation and luteinization [44]. Representative estrous cycles from WT and Gata4+/ females are shown in Figure 1A. The onset of vaginal cornification appeared delayed in Gata4 heterozygotes compared to WT mice (50 ± 6.8 vs. 45 ± 5.5 days, n = 10 per group), although this difference did not reach statistical significance (Fig. 1B). The onset of estrous cyclicity was delayed an average of 12 days in Gata4+/ females compared with WT females (P < 0.01) (Fig. 1B). Average cycle length, however, did not differ between Gata4+/ and WT females (5.3 ± 0.5 and 5.4 ± 0.4 days, respectively), nor did Gata4+/ and WT mice differ in the proportion of time spent in any stage of the estrous cycle (Fig. 1C). The timing of puberty is known to correlate with mouse growth [44]. WT and Gata4+/ female mice had similar growth curves, implying that the delay in puberty in Gata4+/ females was not the result of impaired growth (data not shown). We conclude that Gata4 is a genetic determinant of the onset of puberty in female B6 mice.

FIG. 1.

Delayed puberty in B6 Gata+/− female mice. A) Representative estrous cycles from WT and Gata+/− mice. B) Average ages for first vaginal cornification and the onset of cyclicity. Note the significant delay in the onset of cyclicity in Gata+/− mice. C) Proportion of time spent in each estrous stage (n = 10 for each genotype). Error bars represent SD. D, diestrus; E, estrus; M, metestrus; P, proestrus.

i0006-3363-84-5-1033-f01.tif

The reproductive performance of Gata4+/ female mice was assessed by mating 2-mo-old WT or Gata4+/ female mice with normal males and measuring litter size over successive pregnancies. No differences were found between WT and Gata4+/ mice in either litter size (Table 1) or the frequency of parturition (data not shown).

TABLE 1.

Normal litter size in Gata4+/− female mice.a

i0006-3363-84-5-1033-t01.tif

Ovaries from young adult mice were examined to determine whether Gata4 haploinsufficiency disrupts follicular development. In both WT (Fig. 2, A and B) and Gata4+/ (Fig. 2C) females, nuclear GATA4 immunoreactivity was evident in theca cells and in granulosa cells of primary, preantral, and antral follicles (Fig. 2, A–C), but not in atretic follicles or corpora lutea (Fig. 2, A and C). Consistent with a published report [45], no significant differences were found between WT and Gata4+/ ovaries in the abundance of primordial, primary, or small preantral follicles (healthy or atretic) (Fig. 2, D and E).

FIG. 2.

Ostensibly normal follicular development in the ovaries of randomly cycling adult Gata+/− mice. AC) Ovaries from 3-mo-old WT (A and B) or Gata4+/− (C) mice were subjected to immunoperoxidase staining to assess ovarian morphology and to visualize the distribution of GATA4 within this tissue. Note that GATA4 is expressed in granulosa and theca cells associated with primordial, preantral, and antral follicles of both WT and Gata4+/− mice. Bar = 75 μm. D and E) Histomorphometric analysis of follicle development in WT (n = 10) and Gata4+/− (n = 6) mice. Serial ovarian sections from 7-wk-old WT and Gata4+/− mice were processed, and the numbers of healthy (nonatretic; D) or atretic (E) follicles were estimated. Error bars represent SD. AnF, antral follicles; CL, corpora lutea; PF, primordial and primary follicles; PrF, preantral follicles.

i0006-3363-84-5-1033-f02.tif

Ovaries of Gata4+/− Mice Exhibit an Impaired Response to Exogenous Gonadotropins

Immature WT and Gata4+/ female mice were treated with eCG, which mimics the effect of FSH, followed by hCG, which mimics the effect of LH, to induce synchronized follicular growth and ovulation. Unstimulated WT and Gata4+/ ovaries had similar weights, whereas gonadotropin-stimulated WT ovaries weighed significantly more than their Gata4+/ counterparts (Fig. 3A). Histological analysis confirmed that ovaries of eCG-stimulated WT mice (Fig. 3B) were consistently larger than those of Gata4+/ mice (Fig. 3C).

FIG. 3.

Impaired ovarian response to gonadotropins in Gata+/− mice. A) Weanling WT (solid line) or Gata+/− (dashed line) mice were treated with one of the following regimens: 1) no stimulation; 2) eCG for 48 h; 3) eCG for 48 h, followed by hCG for 18 h; or 4) eCG for 48 h, followed by hCG for 5 days. Ovaries were harvested and weighed (n = 8 for each genotype). *P < 0.05. Error bars represent SD. B and C) Tissue sections of WT (B) and Gata+/− (C) ovaries 48 h after eCG stimulation. The Gata+/− ovary is smaller and contains fewer antral follicles. Bar = 0.3 mm. D) Oocyte yields after gonadotropin-induced ovulation. WT and Gata+/− mice were treated with eCG, followed 48 h later by hCG. Oocytes were harvested from oviducts and counted 16 h after hCG treatment. Each data point represents the oocyte yield of an individual mouse. Horizontal lines represent mean values. E and F) Sections of WT (E) and Gata+/− (F) uteri 48 h after eCG stimulation. Note the hypoestrogenic appearance of the heterozygous uterus. Glandular elements of the endometrial layer are less complex, and only scattered glands are detected in the stroma. No difference was found in mean body weight between the WT and Gata+/− mice (13.8 ± 1.6 and 14.4 ± 1.6 g, respectively). Bar = 0.3 mm. G) Serum E2 levels in WT (white bar, n = 7) and Gata+/− (black bar, n = 7) uteri 48 h after eCG stimulation. *P < 0.05. Error bars represent SD.

i0006-3363-84-5-1033-f03.tif

In response to superovulation, Gata4+/ mice released significantly fewer oocytes into the oviducts than WT mice (Fig. 3D). Thus, Gata4 haploinsufficiency decreases oocyte release in response to exogenous gonadotropins without impairing fertility.

Compared to WT mice, the uteri of eCG-stimulated Gata4+/− mice weighed significantly less (0.32 ± 0.05 [n = 8] vs. 0.20 ± 0.4 [n = 7] mg/g body weight, P < 0.05) and appeared to be hypoestrogenic (Fig. 3, E and F). Following eCG stimulation, serum E2 levels were significantly greater in WT mice than the Gata4+/− mice (Fig. 3G). In contrast, basal serum E2 levels did not differ significantly between WT (8.8 ± 1.1 pg/ml) and Gata4+/ mice (9.5 ± 0.3 pg/ml).

Quantitative RT-PCR was used to measure the relative expression of transcripts for Gata4 (Fig. 4A) and three of its putative target genes, Star (Fig. 4B), Cyp11a1 (Fig. 4C), and Cyp19 (Fig. 4D), in ovaries from gonadotropin-stimulated immature WT and Gata4+/− mice. To confirm a reduction in Gata4 expression in the heterozygous ovaries, we performed qRT-PCR using primers specific for the WT Gata4 mRNA. The ovaries of unstimulated and gonadotropin-stimulated Gata4+/ mice contained approximately half the amount of Gata4 mRNA (normalized to the housekeeping gene L19) as their WT counterparts (Fig. 4A). Basal levels of mRNA for Star (Fig. 4B), Cyp11a1 (Fig. 4C), and Cyp19 (Fig. 4D) were similar in WT and Gata4+/− mice. Following gonadotropin stimulation, however, the levels of Star, Cyp11a1, and Cyp19 mRNA were significantly higher in WT than in Gata4+/ ovaries. The target gene most profoundly affected by Gata4 haploinsufficiency was Cyp19, the key gene of estrogen biosynthesis. Previous studies have shown that CYP19 is essential for follicular growth and coordination of the ovulatory process [4648].

FIG. 4.

Reduced expression of mRNA for Gata4 and steroidogenic factors in the ovaries of gonadotropin-stimulated Gata+/− mice. Weanling WT (solid line, n = 5) or Gata+/− (dashed line, n = 5) mice were treated with one of the following regimens: 1) no stimulation; 2) eCG for 48 h; 3) eCG for 48 h, followed by hCG for 18 h; or 4) eCG for 48 h, followed by hCG for 5 days. Ovarian RNA was subjected to qRT-PCR for Gata4 (A), Star (B), Cyp11a1 (C), and Cyp19 (D). Error bars represent SD. *P < 0.05, **P < 0.01.

i0006-3363-84-5-1033-f04.tif

Conditional Deletion of Gata4 in Granulosa Cells

To further study the physiological function of GATA4 during folliculogenesis, we used the Cre-loxP recombination system to facilitate granulosa cell-specific deletion of the Gata4 gene. We hypothesized that conditional deletion of both Gata4 alleles in granulosa cells would produce a more severe reproductive phenotype than germline deletion of a single allele. The 129;B6 mice homozygous for a floxed allele of Gata4 were intercrossed with 129;B6 Amhr2cre/+ mice. Previous studies have shown that Cre-mediated excision of the region between exons 3 and 5 of the Gata4 gene converts the floxed allele into a recombined allele no longer capable of encoding a functional GATA4 protein [27, 31]. The Amhr2-cre knockin transgene has been widely used to target gene deletion in proliferating granulosa cells and in the mesenchyme-derived cells of the müllerian duct, oviduct, and uterus [33, 4958]. Because GATA4 is not expressed in the müllerian duct (Fig. 5, A and B), oviduct [59], or adult uterus (Fig. 5, C and D), Cre-mediated deletion of the Gata4 gene should not have a direct impact on these tissues.

FIG. 5.

Gata4 mRNA is not expressed in the müllerian duct or adult mouse uterus. Cryosections of Embryonic Day 18.5 mouse embryo (A and B) and Embryonic Day 9.5 gravid uterus (C and D) were subjected to in situ hybridization for GATA4 using radiolabeled riboprobe. Shown are corresponding bright-field (A and C) and dark-field (B and D) photomicrographs. Note that Gata4 mRNA is expressed in fetal ovary (ov) and in endodermal cells of the small intestine (si) and yolk sac (ys), but not in the müllerian duct (md), embryonic ectoderm (em), decidua (de), or uterus (ut). Bar = 100 μm.

i0006-3363-84-5-1033-f05.tif

ROSA26 flox-stop-flox lacZ reporter (R26R) mice were used to assess the expression of Amhr2-cre within ovaries. Consistent with published reports [33, 50, 52], the Amhr2-cre transgene directed expression of Cre to granulosa cells in secondary follicles (Fig. 6A). Interestingly, Cre expression within secondary follicles was not uniform, as evidenced by the presence of both lacZ-positive (Fig. 6A, arrow) and lacZ-negative (Fig. 6A, arrowhead) granulosa cells. Such variegated Cre expression may contribute to the variable penetrance described below.

FIG. 6.

Amhr2-cre-mediated deletion of Gata4Flox in the mouse ovary. A) Amhr2cre/+ mice were crossed with homozygous R26R mice to generate R26R+/−;Amhr2cre/+ mice. Ovaries were cryosectioned and stained with X-gal. Note the variegated pattern of Cre activity in a secondary follicle; both lacZ-positive (arrow) and lacZ-negative (arrowhead) granulosa cells are evident. B) Ovarian RNA was isolated from pairs of 2-mo-old control (Gata4Flox/Flox;Amhr2+/+) or cKO mice and subjected to RT-PCR analysis with primers that distinguish the intact floxed allele from the recombined allele lacking exons 3–5 (Gata4Δex3–5). Note that a transcript derived from recombined allele is seen in the cKO mice but not the control mice. The identity the third band in the cKO mice is unclear. C and D) Ovaries from 7-wk-old Gata4Flox/+;Amhr2cre/+ (C) or cKO (D) mice were sectioned and subjected to immunoperoxidase staining to visualize the distribution of GATA4 within this tissue. GATA4 immunoreactivity was seen in granulosa and theca cells of both the Gata4Flox/+;Amhr2cre/+ and cKO mice. Bar = 75 μm. AAnF, atretic antral follicles; AnF, antral follicles; PF, primordial and primary follicles; PrF, preantral follicles.

i0006-3363-84-5-1033-f06.tif

Intercrosses of Gata4Flox/Flox mice and Gata4Flox/+;Amhr2cre/+ mice yielded (Gata4Flox/Flox;Amhr2cre/+) mice, henceforth referred to as cKO mice, and Gata4Flox/+;Amhr2cre/+ mice in the expected mendelian ratios. The growth of cKO mice did not differ from that of Gata4Flox/+;Amhr2cre/+ mice or Amhr2+/+ controls (data not shown).

To verify Cre-mediated deletion of the Gata4 gene in the mouse ovary, we employed an RT-PCR strategy that distinguishes transcripts derived from the intact (Gata4Flox) and recombined alleles (Gata4Δex3–5). RT-PCR analysis of ovaries from Gata4Flox/Flox;Amhr2+/+ mice yielded a single band corresponding to the intact allele (Fig. 6B). In contrast, analysis of RNA from the ovaries of cKO mice demonstrated both the intact Gata4Flox allele and the recombined Gata4Δex3–5 allele (Fig. 6B). The relative intensities of the Gata4Flox and Gata4Δex3–5 bands differed among cKO mice (Fig. 6B). Variability in the extent of Cre-mediated recombination has been observed in other granulosa cell-specific knockout mice made with Amhr2-cre and may reflect inefficient recombination in granulosa cells (see [52] and references therein).

To confirm reduced expression of GATA4 in the cKO ovaries, we performed qRT-PCR using primers specific for the WT GATA4 mRNA. The ratio of GATA4 mRNA expression in immature cKO versus Gata4Flox/Flox;Amhr2+/+ mice was 0.44 ± 0.02 (P < 0.001). Moreover, the ratio of GATA4 mRNA expression in immature cKO versus Gata4Flox/+;Amhr2cre/+ mice was 0.72 ± 0.02 (P < 0.05).

Next, we compared the patterns of GATA4 immunostaining in ovaries of postpubertal Gata4Flox/+;Amhr2cre/+ (Fig. 6C) and cKO (Fig. 6D) mice. In both genotypes, GATA4 protein was detected in theca cells and in granulosa cells of primary, preantral, and antral follicles. The residual GATA4 expression in the immunohistochemical analysis presumably reflects cells in which the Amhr2-cre did not fully inactivate both Gata4Flox alleles. Nevertheless, we infer from the reproductive defects described below that GATA4 expression is silenced in enough granulosa cells to elicit a phenotype.

Reduced Fertility in Gata4 cKO Mice

To assess the impact of conditional deletion of Gata4 on fertility, female 2-mo-old cKO and Gata4Flox/+;Amhr2cre/+ mice were housed with proven male studs. Both strains gave birth and raised pups to weanlings, but fertility defects were evident in the cKO females. Six of 18 (33%) cKO females failed to produce offspring, compared to only 1 of 20 (5%) Gata4Flox/+;Amhr2cre/+ females (P < 0.05). Even more striking, the average litter size of cKO mice was reduced (P < 0.005) to approximately half that of Gata4Flox/+;Amhr2cre/+ mice (Table 2).

TABLE 2.

Reduced litter size in B6;129 Gata4 cKO mice.

i0006-3363-84-5-1033-t02.tif

Although cKO mice were clearly subfertile, no differences were detected in the onset of puberty or in the length of the estrous cycle among cKO mice, Gata4Flox/+;Amhr2cre/+ mice, and Amhr2+/+ littermate mice (data not shown). That puberty was delayed in B6 Gata4+/− mice (Fig. 1) but not in 129;B6 cKO or Gata4Flox/+;Amhr2cre/+ mice might reflect background strain influences [35] or the timing/efficiency of Cre-mediated ablation. Alternatively, the B6 Gata4+/− mice may have extragonadal defects that delay puberty.

Ovaries of young adult Gata4Flox/+;Amhr2cre/+ (Fig. 7A) and cKO (Fig. 7B) mice were analyzed to determine the impact of conditional ablation of Gata4 on follicular development. On gross inspection, the ovaries of young adult Gata4Flox/+;Amhr2cre/+ and cKO mice were indistinguishable. On histological examination, the number of primordial, primary, and small antral follicles (healthy or atretic) was similar in cKO and Gata4Flox/+;Amhr2cre/+ ovaries (Fig. 7, C and D).

FIG. 7.

Morphological and functional abnormalities in the ovaries of cKO mice. A and B) Hematoxylin-and-eosin-stained sections of ovaries from 7-wk-old Gata4Flox/+;Amhr2cre/+ and cKO mice, respectively. Bar = 200 μm. C and D) Histomorphometric analysis of follicle development in 7-wk-old Gata4Flox/+;Amhr2cre/+ (n = 10) and cKO (n = 6) mice ovaries. Serial ovarian sections were processed, and the numbers of healthy (nonatretic; D) or atretic (E) follicles were estimated. EH) Ovaries from 6-mo-old Gata4Flox/+;Amhr2cre/+ (E and G) and cKO (F and H) mice. The arrowheads and asterisk highlight large cysts in the cKO ovary. Bar = 200 μm. I) Serum AMH levels in Gata4Flox/+;Amhr2cre/+ (n = 3–7) and cKO (n = 5–7) mice of varying ages. Error bars represent SD. The bars denoted by a differ significantly (P < 0.01) from that denoted by b but not from one another.

i0006-3363-84-5-1033-f07.tif

By 6 mo of age, however, differences were found in the gross and microscopic appearance of ovaries from Gata4Flox/+;Amhr2cre/+ (Fig. 7, E and G) and cKO (Fig. 7, F and H) mice. Very large, nonhemorrhagic cysts were seen in six of seven (86%) ovaries from cKO mice and in 0 of 10 (0%) ovaries from Gata4Flox/+;Amhr2cre/+ mice (P < 0.001). The cysts were lined by a flattened epithelium containing scattered ciliated cells (data not shown).

Next, we measured serum AMH levels in Gata4Flox/+;Amhr2cre/+ and cKO mice at different ages (Fig. 7I). Studies of WT mice have shown that serum levels of AMH, a protein secreted by granulosa cells within growing follicles [60], remain relatively constant until 8 mo of age and then decline steadily over the ensuing 10 mo [43]. This age-related decline in serum AMH correlates with decreases in both the number of growing follicles and the size of the primordial follicle pool [43], making AMH a useful marker to quantify ovarian reserve [61]. AMH levels were similar in 2.5-mo-old Gata4Flox/+;Amhr2cre/+ and cKO mice. In contrast, AMH levels were significantly lower (P < 0.01) in 6-mo-old cKO mice. Thus, AMH levels decline prematurely in Gata4 cKO mice, indicating that ovarian granulosa cell function is compromised. In light of these results, we also measured AMH levels in the serum of older B6 WT and Gata4+/− mice but found no significant differences (data not shown).

Gata4 cKO Mice Exhibit an Impaired Response to Exogenous Gonadotropins

Next, we assessed the response of immature Gata4Flox/+;Amhr2cre/+ and cKO mice to superovulation. No significant differences were found in the weight or gross appearance of unstimulated Gata4Flox/+;Amhr2cre/+ and cKO mice (data not shown). However, the ovaries of cKO mice treated with eCG plus hCG were significantly smaller than those of Gata4Flox/+;Amhr2cre/+ mice (4.8 ± 2.0 mg [n = 10] and 7.9 ± 2.0 mg [n = 8], respectively; P < 0.05) and had hemorrhagic follicular cysts on their surface (Fig. 8, A and B). A significant decrease (P < 0.05) was observed in the yield of oocytes from cKO mice compared with Gata4Flox/+;Amhr2cre/+ mice (Fig. 8C), although considerable variability occurred in the response of the cKO mice. One-third of the cKO mice tested released no oocytes following superovulation. Collectively, these data show that the Gata4 granulosa cell-specific knockout mice are subfertile and have an impaired response to gonadotropin-induced superovulation.

FIG. 8.

Aberrant response to superovulation in cKO mice. Immature mice were treated with eCG, followed 48 h later by hCG. Tissue samples were harvested for analysis 18 h after hCG treatment. A and B) Ovaries from eCG/hCG-treated (A) Gata4Flox/+;Amhr2cre/+ and (B) cKO mice. Note the presence of hemorrhagic follicles (arrowheads) in the cKO ovary. C) Oocyte yields after superovulation. Each data point represents the oocyte yield of an individual mouse. The horizontal lines represent the mean values. The sample sizes were Gata4Flox/+;Amhr2cre/+ (n = 11) and cKO (n = 13).

i0006-3363-84-5-1033-f08.tif

Quantitative RT-PCR was used to measure the relative expression of transcripts for Gata4 (Fig. 9A) and its target genes Star (Fig. 9B), Cyp11a1 (Fig. 9C), and Cyp19 (Fig. 9D) in ovaries from gonadotropin-stimulated, immature Gata4Flox/+;Amhr2cre/+ and cKO mice. Basal levels of mRNA for Star, Cyp11a1, and Cyp19 were similar in Gata4Flox/+;Amhr2cre/+ and cKO mice. Following eCG stimulation, however, the level of Cyp19 mRNA was significantly higher in Gata4Flox/+;Amhr2cre/+ than in cKO ovaries. Additionally, a trend, albeit statistically insignificant, was observed toward decreased expression of Star and Cyp11a1 in the ovaries of gonadotropin-stimulated cKO mice. We did not, however, detect a significant difference in the serum E2 levels of eCG-treated juvenile mice.

FIG. 9.

Reduced expression of mRNA for Gata4 and steroidogenic factors in the ovaries of gonadotropin-stimulated cKO mice. Weanling Gata4Flox/+;Amhr2cre/+ (solid line, n = 5) or cKO (dashed line, n = 5) mice were treated with one of the following regimens: 1) no stimulation; 2) eCG for 48 h; 3) eCG for 48 h, followed by hCG for 18 h; or 4) eCG for 48 h, followed by hCG for 5 days. Ovarian RNA was subjected to qRT-PCR for Gata4 (A), Star (B), Cyp11a1 (C), and Cyp19 (D). Error bars represent SD. *P < 0.05, **P < 0.01.

i0006-3363-84-5-1033-f09.tif

DISCUSSION

Expression of GATA4 occurs in potentially mitotic and proliferating granulosa cells, but expression is lost once these cells become either terminally differentiated (from granulosa to luteal cells) or apoptotic during follicular atresia [4, 7]. On the basis of these expression patterns, it was suggested [4] that GATA4 might control genes involved in the maintenance or maturation of granulosa cells within early follicles (i.e., before ovulation). Consistent with its proposed role in granulosa cell maturation, the expression level of GATA4 in granulosa cells was subsequently shown to be enhanced by FSH, and the transcriptional activity of GATA4 was found to be induced by phosphorylation via signaling through the FSH receptor and the downstream mediators cAMP and protein kinase A [8, 9]. The abrupt decrease in GATA4 associated with ovulation or apoptosis led investigators to propose that this factor is not required for the later stages of follicular development, apoptosis, or luteinization, although GATA4 might serve to prime early follicular cells for the transition to late maturation or apoptosis [4]. In keeping with these previous observations and predictions, the present data demonstrate that germline and conditional deletions in the Gata4 gene cause defects in granulosa cell function that impair ovarian function in adult mice.

The B6 Gata4+/− female mice were fertile but had delayed puberty and a blunted ovarian response to exogenous gonadotropins; the ovaries of gonadotropin-stimulated Gata4+/− mice were smaller, released fewer oocytes, and expressed less mRNA for the steroidogenic genes Star, Cyp11a1, and Cyp19 (Table 3). In the absence of a fertility defect, the physiological significance of a reduced response to ovarian superstimulation is arguable. On the other hand, our data show that provocative stimulation can be used to highlight “subclinical” defects in hormonal responsiveness that shed light on the function of GATA4 in vivo. The steroidogenic gene most profoundly affected by Gata4 haploinsufficiency was Cyp19, the key gene of estrogen biosynthesis and a known target for activation by GATA4 [8, 9]. Consistent with reduced Cyp19 expression, serum E2 levels were significantly reduced in Gata4+/− mice (Fig. 3G), and the uteri of eCG-stimulated Gata4+/− mice appeared to be hypoestrogenic. Like Gata4+/− mice, Cyp19 knockout mice have impaired ovulation and hypoestrogenic uteri [4648], suggesting that the abnormal phenotype of gonadotropin-stimulated Gata4 haploinsufficient mice may reflect, at least in part, attenuated Cyp19 expression and a concomitant decrease in E2 production. We speculate that the delay in the onset of puberty in Gata4+/− female mice could reflect a slight delay in the production of E2 in these mice, although we have no direct evidence of this and cannot exclude an extragonadal effect of Gata4 haploinsufficiency. That Gata4+/− female mice have a normal estrous cycle length suggests that these animals eventually achieve adequate plasma levels of E2.

TABLE 3.

Comparison of the phenotypes of the two models of GATA4 deficiency used in this study.

i0006-3363-84-5-1033-t03.tif

The Gata4 cKO females have a more profound phenotype, including impaired fertility, cystic ovarian changes, and an attenuated ovulation response to exogenous gonadotropins (Table 3). As with the Gata4 haploinsufficient mice, the steroidogenic gene most profoundly affected by conditional deletion of the Gata4 gene was Cyp19. That most cKO mice were subfertile rather than completely infertile might reflect an inherent limitation of using Amhr2-cre to drive tissue-specific recombination. As shown by studies of β-catenin cKO mice generated using Amhr2-cre [52], the importance of a given gene for ovarian function may be masked by compensatory responses that promote selective proliferation of granulosa cells in follicles that escape Cre-mediated recombination. Indeed, a hallmark of the Gata4 cKO mice and other knockout mice generated using Amhr2-cre is animal-to-animal variability, which may reflect inefficient recombination in granulosa cells (see [52] and references therein).

In Gata4 cKO, but not Gata4+/−, mice, serum AMH levels declined earlier than in age-matched controls. Studies have shown that AMH, which is produced by granulosa cells in growing follicles, suppresses the development of primordial follicles [6264]. In the absence of AMH, the primordial follicle pool depletes prematurely [62]. Serum AMH levels correlate with the size of the primordial follicle pool, making it a useful serum marker of ovarian reserve in both mice and women [61]. Of note, the Amh gene has been shown to be a direct target for activation of GATA4 in the testis and ovary (see [1] and references therein). Thus, it remains unclear whether the decrease in serum AMH in the cKO mice reflects a decrease in ovarian reserve, reduced GATA4-dependent activation of the Amh promoter, or both. Regardless of the mechanism, the premature decline in AMH levels underscores that ovarian granulosa cell function is compromised in the cKO mice.

Studies by Tevosian and colleagues [3, 19, 22] have established that GATA4 and its cofactor, ZFPM2, mediate early events in ovarian development. In particular, these transcriptional regulators appear to modulate the response of embryonic ovarian tissue to extracellular signals, such as the WNT/β-catenin pathway. We presume, based on the appearance of unstimulated ovaries from young mice, that fetal ovarian development proceeds normally in both Gata4 haploinsufficient mice and granulosa cell-specific cKO mice. This is in accordance with the results of studies on combined haploinsufficiency of Nr5a1 and Gata4, which did not reveal a genetic interaction in mouse gonadal development [45]. In the case of Gata4 cKO mice, we speculate that fetal ovarian development is normal, because efficient Cre-mediated recombination does not take place until robust expression of Amhr2-cre occurs in secondary and small antral follicles [52, 65].

In summary, the present study establishes a role for GATA4 in the regulation of adult ovarian function. These results, coupled with those of other investigators [9], show that GATA4 regulates the response of postnatal ovarian tissue to gonadotropins. Similarities in the postnatal ovarian phenotypes of Gata4 cKO mice and mice harboring granulosa cell-specific knockouts of genes in the WNT/β-catenin pathway [52, 57] raise the possibility that GATA4 regulates this signaling pathway not only in fetal mice [3, 19, 22] but also in adults; further experiments will explore this possibility. In a complementary study [66], we show that GATA4 also regulates testicular function in adults; male mice in which Gata4 was conditionally deleted in Sertoli cells using Amhr2-cre exhibit impaired fertility and age-dependent testicular atrophy.

Acknowledgements

We thank Christine Ratajczak for technical advice and members of the Digestive Diseases Research Center Histology Core for their assistance. We also thank Simone Wagner for assistance with the Gata4+/ experiments. The Gata4+/− mice were generously provided by William Pu (Children's Hospital, Boston, MA).

REFERENCES

1.

RS Viger, SM Guittot, M Anttonen, DB Wilson, and M Heikinheimo . Role of the GATA family of transcription factors in endocrine development, function, and disease. Mol Endocrinol 2008. 22:781–798. Google Scholar

2.

HA LaVoie . The role of GATA in mammalian reproduction. Exp Biol Med 2003. 228:1282–1290. Google Scholar

3.

SG Tevosian and NL Manuylov . To beta or not to beta: canonical beta-catenin signaling pathway and ovarian development. Dev Dyn 2008. 237:3672–3680. Google Scholar

4.

M Heikinheimo, M Ermolaeva, M Bielinska, NA Rahman, N Narita, IT Huhtaniemi, JS Tapanainen, and DB Wilson . Expression and hormonal regulation of transcription factors GATA-4 and GATA-6 in the mouse ovary. Endocrinology 1997. 138:3505–3514. Google Scholar

5.

HA LaVoie, GL McCoy, and CA Blake . Expression of the GATA-4 and GATA-6 transcription factors in the fetal rat gonad and in the ovary during postnatal development and pregnancy. Mol Cell Endocrinol 2004. 227:31–40. Google Scholar

6.

SA McCoard, TH Wise, SC Fahrenkrug, and JJ Ford . Temporal and spatial localization patterns of Gata4 during porcine gonadogenesis. Biol Reprod 2001. 65:366–374. Google Scholar

7.

MP Laitinen, M Anttonen, I Ketola, DB Wilson, O Ritvos, R Butzow, and M Heikinheimo . Transcription factors GATA-4 and GATA-6 and a GATA family cofactor, FOG-2, are expressed in human ovary and sex cord-derived ovarian tumors. J Clin Endocrinol Metab 2000. 85:3476–3483. Google Scholar

8.

HA LaVoie, D Singh, and YY Hui . Concerted regulation of the porcine steroidogenic acute regulatory protein gene promoter activity by follicle-stimulating hormone and insulin-like growth factor I in granulosa cells involves GATA-4 and CCAAT/enhancer binding protein beta. Endocrinology 2004. 145:3122–3134. Google Scholar

9.

J Kwintkiewicz, Z Cai, and C Stocco . Follicle-stimulating hormone-induced activation of Gata4 contributes in the up-regulation of Cyp19 expression in rat granulosa cells. Mol Endocrinol 2007. 21:933–947. Google Scholar

10.

A Balla, N Danilovich, Y Yang, and MR Sairam . Dynamics of ovarian development in the FORKO immature mouse: structural and functional implications for ovarian reserve. Biol Reprod 2003. 69:1281–1293. Google Scholar

11.

TE Vaskivuo, K Aittomaki, M Anttonen, A Ruokonen, R Herva, Y Osawa, M Heikinheimo, I Huhtaniemi, and JS Tapanainen . Effects of follicle-stimulating hormone (FSH) and human chorionic gonadotropin in individuals with an inactivating mutation of the FSH receptor. Fertil Steril 2002. 78:108–113. Google Scholar

12.

H Hiroi, LK Christenson, and JF Strauss III. . Regulation of transcription of the steroidogenic acute regulatory protein (StAR) gene: temporal and spatial changes in transcription factor binding and histone modification. Mol Cell Endocrinol 2004. 215:119–126. Google Scholar

13.

YY Hui and HA Lavoie . GATA4 reduction enhances 3′,5′-cyclic adenosine 5′-monophosphate-stimulated steroidogenic acute regulatory protein messenger ribonucleic acid and progesterone production in luteinized porcine granulosa cells. Endocrinology 2008. 149:5557–5567. Google Scholar

14.

N Sher, N Yivgi-Ohana, and J Orly . Transcriptional regulation of the cholesterol side chain cleavage cytochrome P450 gene (CYP11A1) revisited: binding of GATA, cyclic adenosine 3′,5′-monophosphate response element-binding protein and activating protein (AP)-1 proteins to a distal novel cluster of cis-regulatory elements potentiates AP-2 and steroidogenic factor-1-dependent gene expression in the rodent placenta and ovary. Mol Endocrinol 2007. 21:948–962. Google Scholar

15.

JJ Tremblay and RS Viger . GATA factors differentially activate multiple gonadal promoters through conserved GATA regulatory elements. Endocrinology 2001. 142:977–986. Google Scholar

16.

MF Bouchard, H Taniguchi, and RS Viger . Protein kinase A-dependent synergism between GATA factors and the nuclear receptor, liver receptor homolog-1, regulates human aromatase (CYP19) PII promoter activity in breast cancer cells. Endocrinology 2005. 146:4905–4916. Google Scholar

17.

C Stocco . In vivo and in vitro inhibition of cyp19 gene expression by prostaglandin F2α in murine luteal cells: implication of GATA-4. Endocrinology 2004. 145:4957–4966. Google Scholar

18.

C Stocco, J Kwintkiewicz, and Z Cai . Identification of regulatory elements in the Cyp19 proximal promoter in rat luteal cells. J Mol Endocrinol 2007. 39:211–221. Google Scholar

19.

NL Manuylov, FO Smagulova, L Leach, and SG Tevosian . Ovarian development in mice requires the GATA4-FOG2 transcription complex. Development 2008. 135:3731–3743. Google Scholar

20.

S Vainio, M Heikkila, A Kispert, N Chin, and AP McMahon . Female development in mammals is regulated by Wnt-4 signaling. Nature 1999. 397:405–409. Google Scholar

21.

NH Uhlenhaut and M Treier . Foxl2 function in ovarian development. Mol Genet Metab 2006. 88:225–234. Google Scholar

22.

SG Tevosian, KH Albrecht, JD Crispino, Y Fujiwara, EM Eicher, and SH Orkin . Gonadal differentiation, sex determination and normal Sry expression in mice require direct interaction between transcription partners GATA4 and FOG2. Development 2002. 129:4627–4634. Google Scholar

23.

CT Kuo, EE Morrisey, R Anadappa, K Sigrist, MM Lu, MS Parmacek, C Soudais, and JM Leiden . GATA4 transcription factor is required for ventral morphogenesis and heart tube formation. Genes Dev 1997. 11:1048–1060. Google Scholar

24.

JD Molkentin, Q Lin, SA Duncan, and EN Olson . Requirement of the transcription factor GATA4 for heart tube formation and ventral morphogenesis. Genes Dev 1997. 11:1061–1072. Google Scholar

25.

N Narita, M Bielinska, and D Wilson . Cardiomyocyte differentiation by GATA-4 deficient embryonic stem cells. Devel . 1997. 122:3755–3764. Google Scholar

26.

N Narita, M Bielinska, and DB Wilson . Wild-type endoderm abrogates the ventral developmental defects associated with GATA-4 deficiency in the mouse. Dev Biol 1997. 189:270–274. Google Scholar

27.

AJ Watt, MA Battle, J Li, and SA Duncan . GATA4 is essential for formation of the proepicardium and regulates cardiogenesis. Proc Natl Acad Sci U S A 2004. 101:12573–12578. Google Scholar

28.

B Thurisch, S Liang, N Sarioglu, L Schomburg, J Bungert, and C Dame . Transgenic mice expressing small interfering RNA against Gata4 point to a crucial role of Gata4 in the heart and gonads. J Mol Endocrinol 2009. 43:157–169. Google Scholar

29.

WT Pu, T Ishiwata, AL Juraszek, Q Ma, and S Izumo . GATA4 is a dosage-sensitive regulator of cardiac morphogenesis. Dev Biol 2004. 275:235–244. Google Scholar

30.

EM Zeisberg, Q Ma, AL Juraszek, K Moses, RJ Schwartz, S Izumo, and WT Pu . Morphogenesis of the right ventricle requires myocardial expression of Gata4. J Clin Invest 2005. 115:1522–1531. Google Scholar

31.

T Oka, M Maillet, AJ Watt, RJ Schwartz, BJ Aronow, SA Duncan, and JD Molkentin . Cardiac-specific deletion of Gata4 reveals its requirement for hypertrophy, compensation, and myocyte viability. Circ Res 2006. 98:837–845. Google Scholar

32.

SP Jamin, NA Arango, Y Mishina, MC Hanks, and RR Behringer . Requirement of Bmpr1a for mullerian duct regression during male sexual development. Nat Genet 2002. 32:408–410. Google Scholar

33.

P Jeyasuria, Y Ikeda, SP Jamin, L Zhao, DG de Rooij, AP Themmen, RR Behringer, and KL Parker . Cell-specific knockout of steroidogenic factor 1 reveals its essential roles in gonadal function. Mol Endocrinol 2004. 18:1610–1619. Google Scholar

34.

G Friedrich and P Soriano . Promoter traps in embryonic stem cells: a genetic screen to identify and mutate developmental genes in mice. Genes Dev 1991. 5:1513–1523. Google Scholar

35.

JF Nelson, LS Felicio, PK Randall, C Sims, and CE Finch . A longitudinal study of estrous cyclicity in aging C57BL/6J mice: I. Cycle frequency, length and vaginal cytology. Biol Reprod 1982. 27:327–339. Google Scholar

36.

PY Jay, M Bielinska, JM Erlich, S Mannisto, WT Pu, M Heikinheimo, and DB Wilson . Impaired mesenchymal cell funciton in Gata4 mutant mice leads to diaphragmatic hernias and primary lung defects. Dev Biol 2007. 301:602–614. Google Scholar

37.

M Bielinska, PY Jay, JM Erlich, S Mannisto, Z Urban, M Heikinheimo, and DB Wilson . Molecular genetics of congenital diaphragmatic defects. Ann Med 2007. 39:261–274. Google Scholar

38.

SK Rajagopal, Q Ma, D Obler, J Shen, A Manichaikul, A Tomita-Mitchell, K Boardman, C Briggs, V Garg, D Srivastava, E Goldmuntz, KW Broman, et al . Spectrum of heart disease associated with murine and human GATA4 mutation. J Mol Cell Cardiol 2007. 43:677–685. Google Scholar

39.

Y Morita, GI Perez, DV Maravei, KI Tilly, and JL Tilly . Targeted expression of Bcl-2 in mouse oocytes inhibits ovarian follicle atresia and prevents spontaneous and chemotherapy-induced oocyte apoptosis in vitro. Mol Endocrinol 1999. 13:841–850. Google Scholar

40.

JL Tilly . Ovarian follicle counts—not as simple as 1, 2, 3. Reprod Biol Endocrinol 2003. 1:11–15. Google Scholar

41.

B Hogan, R Beddington, F Costantini, and E Lacy . Manipulating the Mouse Embryo: A Laboratory Manual. . Cold Spring Harbor, NY: Cold Spring Harbor Press;. 1994. .  Google Scholar

42.

A Kyronlahti, M Ramo, M Tamminen, L Unkila-Kallio, R Butzow, A Leminen, M Nemer, N Rahman, I Huhtaniemi, M Heikinheimo, and M Anttonen . GATA-4 regulates Bcl-2 expression in ovarian granulosa cell tumors. Endocrinology 2008. 149:5635–5642. Google Scholar

43.

ME Kevenaar, MF Meerasahib, P Kramer, BM van de Lang-Born, FH de Jong, NP Groome, AP Themmen, and JA Visser . Serum anti-mullerian hormone levels reflect the size of the primordial follicle pool in mice. Endocrinology 2006. 147:3228–3234. Google Scholar

44.

JF Nelson, K Karelus, LS Felicio, and TE Johnson . Genetic influences on the timing of puberty in mice. Biol Reprod 1990. 42:649–655. Google Scholar

45.

C Pelusi, L Zhao, NR Stallings, and KL Parker . Combined haploinsufficiency of SF-1 and GATA4 does not reveal a genetic interaction in mouse gonadal development. Sex Dev 2007. 1:152–160. Google Scholar

46.

CR Fisher, KH Graves, AF Parlow, and ER Simpson . Characterization of mice deficient in aromatase (ArKO) because of targeted disruption of the cyp19 gene. Proc Natl Acad Sci U S A 1998. 95:6965–6970. Google Scholar

47.

KL Britt, AE Drummond, VA Cox, M Dyson, NG Wreford, ME Jones, ER Simpson, and JK Findlay . An age-related ovarian phenotype in mice with targeted disruption of the Cyp19 (aromatase) gene. Endocrinology 2000. 141:2614–2623. Google Scholar

48.

K Toda, K Takeda, T Okada, S Akira, T Saibara, T Kaname, K Yamamura, S Onishi, and Y Shizuta . Targeted disruption of the aromatase P450 gene (Cyp19) in mice and their ovarian and uterine responses to 17beta-estradiol. J Endocrinol 2001. 170:99–111. Google Scholar

49.

E Deutscher and H Hung-Chang Yao . Essential roles of mesenchyme-derived beta-catenin in mouse mullerian duct morphogenesis. Dev Biol 2007. 307:227–236. Google Scholar

50.

G Gonzalez and RR Behringer . Dicer is required for female reproductive tract development and fertility in the mouse. Mol Reprod Dev 2009. 76:678–688. Google Scholar

51.

GD Orvis, SP Jamin, KM Kwan, Y Mishina, VM Kaartinen, S Huang, AB Roberts, L Umans, D Huylebroeck, A Zwijsen, D Wang, JF Martin, et al . Functional redundancy of TGF-beta family type I receptors and receptor-Smads in mediating anti-mullerian hormone-induced mullerian duct regression in the mouse. Biol Reprod 2008. 78:994–1001. Google Scholar

52.

JA Hernandez Gifford, ME Hunzicker-Dunn, and JH Nilson . Conditional deletion of beta-catenin mediated by Amhr2cre in mice causes female infertility. Biol Reprod 2009. 80:1282–1292. Google Scholar

53.

FG Petit, SP Jamin, I Kurihara, RR Behringer, FJ DeMayo, MJ Tsai, and SY Tsai . Deletion of the orphan nuclear receptor COUP-TFII in uterus leads to placental deficiency. Proc Natl Acad Sci U S A 2007. 104:6293–6298. Google Scholar

54.

HY Fan, M Shimada, Z Liu, N Cahill, N Noma, Y Wu, J Gossen, and JS Richards . Selective expression of KrasG12D in granulosa cells of the mouse ovary causes defects in follicle development and ovulation. Development 2008. 135:2127–2137. Google Scholar

55.

R Duggavathi, DH Volle, C Mataki, MC Antal, N Messaddeq, J Auwerx, BD Murphy, and K Schoonjans . Liver receptor homolog 1 is essential for ovulation. Genes Dev 2008. 22:1871–1876. Google Scholar

56.

Y Ren, RG Cowan, RM Harman, and SM Quirk . Dominant activation of the hedgehog signaling pathway in the ovary alters theca development and prevents ovulation. Mol Endocrinol 2009. 23:711–723. Google Scholar

57.

A Boyer, E Lapointe, X Zheng, RG Cowan, H Li, SM Quirk, FJ Demayo, JS Richards, and D Boerboom . WNT4 is required for normal ovarian follicle development and female fertility. FASEB J 2010. 24:3010–3025. Google Scholar

58.

C Pelusi, Y Ikeda, M Zubair, and KL Parker . Impaired follicle development and infertility in female mice lacking steroidogenic factor 1 in ovarian granulosa cells. Biol Reprod 2008. 79:1074–1083. Google Scholar

59.

M Anttonen, I Ketola, H Parviainen, AK Pusa, and M Heikinheimo . FOG-2 and GATA-4 are coexpressed in the mouse ovary and can modulate mullerian-inhibiting substance expression. Biol Reprod 2003. 68:1333–1340. Google Scholar

60.

A Munsterberg and R Lovell-Badge . Expression of the mouse anti-mullerian hormone gene suggests a role in both male and female sexual differentiation. Development 1991. 113:613–624. Google Scholar

61.

EL van Houten, AP Themmen, and JA Visser . Anti-mullerian hormone (AMH): regulator and marker of ovarian function. Ann Endocrinol (Paris ) 2010. 71:191–197. Google Scholar

62.

MJ Gruijters, JA Visser, AL Durlinger, and AP Themmen . Anti-mullerian hormone and its role in ovarian function. Mol Cell Endocrinol 2003. 211:85–90. Google Scholar

63.

C Weenen, JS Laven, AR Von Bergh, M Cranfield, NP Groome, JA Visser, P Kramer, BC Fauser, and AP Themmen . Anti-mullerian hormone expression pattern in the human ovary: potential implications for initial and cyclic follicle recruitment. Mol Hum Reprod 2004. 10:77–83. Google Scholar

64.

AL Durlinger, MJ Gruijters, P Kramer, B Karels, HA Ingraham, MW Nachtigal, JT Uilenbroek, JA Grootegoed, and AP Themmen . Anti-mullerian hormone inhibits initiation of primordial follicle growth in the mouse ovary. Endocrinology 2002. 143:1076–1084. Google Scholar

65.

CJ Jorgez, M Klysik, SP Jamin, RR Behringer, and MM Matzuk . Granulosa cell-specific inactivation of follistatin causes female fertility defects. Mol Endocrinol 2004. 18:953–967. Google Scholar

66.

A Kyronlahti, R Euler, M Bielinska, EL Schoeller, KH Moley, J Toppari, M Heikinheimo, and DB Wilson . GATA4 regulates Sertoli cell function and fertility in adult male mice. Mol Cell Endocrinol 2011. 333:85–95. Google Scholar

Notes

[1] Financial disclosure Supported by NIH DK075618 and DK52574, Academy of Finland, and the Sigrid Juselius Foundation.

[2] Equal contributor 3These authors contributed equally to the work.

Antti Kyrönlahti, Melanie Vetter, Rosemarie Euler, Malgorzata Bielinska, Patrick Y. Jay, Mikko Anttonen, Markku Heikinheimo, and David B. Wilson "GATA4 Deficiency Impairs Ovarian Function in Adult Mice," Biology of Reproduction 84(5), 1033-1044, (19 January 2011). https://doi.org/10.1095/biolreprod.110.086850
Received: 21 June 2010; Accepted: 1 January 2011; Published: 19 January 2011
KEYWORDS
developmental biology
fertility
gene regulation
granulosa cells
ovary
ovulation
puberty
Back to Top