The Role of Gonadotropin-Releasing Hormone in Murine ...

2 downloads 30 Views 974KB Size Report
embryos were cultured with GnRH agonist and antagonist in vitro to assess the influence of GnRH analogs on embryo development. GnRH is expressed in the ...
0013-7227/99/$03.00/0 Endocrinology Copyright © 1999 by The Endocrine Society

Vol. 140, No. 8 Printed in U.S.A.

The Role of Gonadotropin-Releasing Hormone in Murine Preimplantation Embryonic Development* ˜ †, JAN KRUESSEL, YAN WEN, FRANCISCO RAGA†, EVA MARIA CASAN FERNANDO BONILLA-MUSOLES, AND MARY LAKE POLAN Department of Gynecology and Obstetrics, Reproductive Immunology Laboratory, Stanford University School of Medicine (F.R., E.M.C., J.K., Y.W., M.L.P.), Stanford, California; and the Department of Obstetrics and Gynecology, University of Valencia School of Medicine (F.R., E.M.C., F.B.-M.), and the Center for Gynecology and Obstetrics (F.R., E.M.C., F.B.-M.), 46004 Valencia, Spain ABSTRACT Previous studies have established the presence of an extrahypothalamic GnRH in a variety of tissues. GnRH receptor is known to be present in the placenta, which produces and secretes the decapeptide from the very early stages of placentation. We hypothesized that GnRH may play a role in the preimplantation development of embryos. To examine this hypothesis, we assessed GnRH and GnRH receptor messenger RNA (mRNA; RT-PCR) and protein expression (Immunohistochemistry) in preimplantation murine embryos at various developmental stages. Furthermore, preimplantation murine embryos were cultured with GnRH agonist and antagonist in vitro to assess the influence of GnRH analogs on embryo development.

G

nRH PLAYS a crucial role in the regulation of reproductive function. This decapeptide is released from the hypothalamus and binds to specific receptors on the anterior pituitary, resulting in stimulation of both synthesis and release of FSH and LH (1). In addition to the central action, a variety of tissues in several mammalian species express an extrahypothalamic GnRH that is immunologically, biologically, and chemically identical to the hypothalamic hormone (2–7). Moreover, the presence of low affinity/high capacity binding sites for GnRH has been demonstrated in several extrapituitary organs such as the placenta, endometrium, myometrium, breast, prostate, ovary, and testis (5, 7, 8). Preimplantation embryonic development and implantation are a complex series of steps that under normal circumstances begin even before the blastocyst reaches the uterine cavity and attaches to the endometrial epithelium after loss of the zona pellucida. To complete this enigmatic process, there is an embryonic-maternal dialogue, in which the embryo and the endometrium induce changes in each other to promote embryonic development and endometrial receptivity (9, 10). Cytokines, growth factors, and their receptors have been Received July 31, 1998. Address all correspondence and requests for reprints to: Francisco Raga, M.D., Center for Gynecology and Obstetrics, Navarro Reverter 11–1, 46004 Valencia, Spain. E-mail: [email protected]. * This work was supported in part by NIH Grant HD-31575 (to M.L.P.). † Postdoctoral research fellow supported by Ministerio de Sanidad y Consumo grant (FIS 97/5374 and 99/0657) from the Spanish Government (Madrid, Spain).

GnRH is expressed in the developing mouse embryo from morula to hatching blastocyst stages at the mRNA and protein levels. GnRH receptor mRNA is also present in the developing embryos studied. Preimplantation embryonic development was significantly enhanced by incubation with increasing concentrations of GnRH agonist and is significantly decreased by GnRH antagonist compared with that in the control group. Moreover, GnRH antagonist (5 and 10 mM) was able to completely block embryo development. The deleterious effect of GnRH antagonist on embryo development was reversed by increasing concentrations of the agonist, as determined by the number of embryos reaching the blastocyst stage. (Endocrinology 140: 3705–3712, 1999)

detected in pre- and periimplantation embryos, the fallopian tubes, and uterine endometrium (10 –13). Their role in embryo development, endometrial preparation, and the implantation process has been implicated. For instance, preimplantation embryos cultured in vitro lag in development compared with their in vivo counterparts (10). Preimplantation embryonic development improves when the embryos are cocultured with other cells, such as endometrial epithelium and Vero cells (14, 15). These observations suggest that although embryos can develop successfully in vitro, the maternal reproductive tract is likely to provide other factors that further enhance embryo development and implantation. Recent studies from our group demonstrated the presence of GnRH and its receptor at both messenger RNA (mRNA) and protein levels in human endometrium of fertile patients with a dynamic pattern, showing a rise in the midluteal phase, the time of embryonic implantation (6, 7). Furthermore, the presence of a GnRH receptor on the murine uterus and its possible paracrine role in endometrial dynamics have been elucidated (13, 16). Moreover, several reports concerning inadvertent exposure to GnRH agonist of human pregnancies during the early stages of embryonic development and implantation have been reported in in vitro fetilization patients. The clinical experience in those cases has suggested that not only are detrimental effects unlikely, but also the analog might enhance early embryonic development and implantation (17, 18). We hypothesized that an interaction between the embryo and maternal reproductive tract via the GnRH system may be playing an important role during preimplantation embryonic development and the implantation process.

3705

3706

Endo • 1999 Vol 140 • No 8

GnRH IN MURINE EMBRYONAL DEVELOPMENT

Materials and Methods Animals preparation and embryo recovery The planning and conduct of the experimental procedures as well as the maintenance of the animals were carried out in accordance with the NIH Guide for the Care and Use of Laboratory Animals. B6C3F1 mice, 12 weeks of age, were obtained from Charles River Laboratories, Inc. (Wilmington, MA) and maintained at 22–24 C on a 12-h light, 12-h dark cycle. Female mice were superovulated by ip injection of 10 IU PMSG (Sigma Chemical Co. St. Louis, MO) and injected 48 h later with 10 IU hCG (Sigma Chemical Co.) to induce ovulation. Female mice were mated with male mice of the same strain and age; a single male was placed with two females overnight. Mating was evidenced by the appearance of a vaginal plug, and mice were killed by cervical dislocation 22 h post-hCG administration, following a protocol approved by the committee on animal care and use at Stanford University (Palo Alto, CA). Ovaries and oviducts were removed and washed in modified Ham’s F-10 medium (Irvine Scientific, Santa Ana, CA). Cumulus complexes were flushed from the oviducts under microscopic control. Cumuli were removed by incubation in 80 IU/ml hyaluronidase (Sigma Chemical Co.) for 3 min and gentle suction through a Pasteur pipette (Fisher Scientific, Santa Ana, CA) pulled to an inner diameter of approximately 200 mm. Cumulus-free zygotes were washed in medium twice to remove hyaluronidase and transferred to 35-mm culture dishes (Falcon, Becton, Dickinson and Co., Lincoln Park, NJ) containing four single drops of 20 ml modified Ham’s F-10 medium containing 0.3% BSA (Sigma Chemical Co.), covered with light white mineral oil (Sigma Chemical Co.) (19). Medium, hyaluronidase, and oil were equilibrated for 24 h before use in a tissue culture incubator (Forma Scientific, Marietta, OH) at 37 C with a humidified atmosphere of 95% air and 5% CO2. Embryos were cultured under the same atmospheric conditions and were checked the following morning to monitor fertilization. Two-cell embryos were removed, washed with fresh medium, and then randomly transferred to drops of modified Ham’s F-10 containing 0.3% BSA (Sigma Chemical Co.) (19). Embryos were checked daily by inverted microscope (Olympus Corp., Tokyo, Japan) for 3 consecutive days.

RT-PCR analysis Single embryos from 88 h post-hCG (compacted morula; n 5 60), 95 h post-hCG (early expanded blastocyst; n 5 60), and 120 h post-hCG (hatching blastocyst; n 5 60) were examined by RT followed by two rounds of nested PCR using a modification of methods described previously (14, 20) for b-actin (internal standard), GnRH, and GnRH receptor mRNAs. Sequences of complementary DNA (cDNA) clones for the mRNAs that should be detected in single embryos; b-actin (21), GnRH (22), and GnRH receptor (23) were obtained from the GenBank database of the National Center for Biotechnology Information (internet address: http://www.2.ncbi.nlm.nih.gov/cgi-bin/GenBank). The corresponding primer sequences were constructed with the help of the program OLIGO 5.0 Primer Analysis Software (National Bioscience, Plymouth, MN) and synthesized by the Protein, Amino Acid, and Nucleic Acid Facility (Stanford University Medical Center). To ensure that the product detected resulted from amplification of cDNA rather than

contaminating genomic DNA, primers were designed to cross intron/ exon boundaries. As a negative control for specific primers, a defined volume of culture medium in which the embryos were cultured was subjected to the same RT-PCR reaction. The primer cDNA sequences and the sizes of the amplified fragments are listed in Table 1. To detect the mRNA transcripts of single embryos with RT-PCR, the RT was performed with oligo(deoxythymidine)16 to transcribe all of the cDNA fragments to be studied. For the specific primer mixture used in the PCR, 39- and 59-primers of each specific outer pair for the first PCR and 39- and 59-primers of each specific inner pair for the second PCR were mixed to a final concentration of 5 mm. RT Mastermix for each embryo was prepared containing 5 mm MgCl2, 10 3 PCR buffer II, 1 mm of each deoxy (d)-NTP (Perkin Elmer, Foster City, CA), and 2.5 mm oligo(deoxythymidine)16 and placed in a 0.5-ml thin walled PCR tube (Applied Scientific, San Francisco, CA). RT Mastermix in each PCR tube was covered with 50 ml light white oil (Sigma Chemical Co.) and kept on ice until embryo collection. A single embryo carrying about 1 ml culture medium was aspirated with a micropipette and transferred to the PCR tube containing 17.5 ml RT Mastermix. Samples were immediately heated to 99 C for 1 min in a DNA thermal cycler 480 (Perkin Elmer) to release the total RNA and denature the proteins. Samples were cooled to 4 C, and 20 U ribonuclease inhibitor (Perkin Elmer) and 100 U Moloney murine leukemia virus reverse transcriptase (Life Technologies, Grand Island, NY) were added before starting the RT. The RT reaction in a total volume of 20 ml was carried out in the DNA thermal cycler 480 using a program with one 30-min RT cycle at 42 C, followed by 5 min at 99 C, then quenching at 4 C. Products were stored at 220 C until the subsequent PCR. For the first PCR, 2 ml RT product from an individual embryo were added to the first PCR Mastermix to a total volume of 49.5 ml containing 2 mm MgCl2, 10 3 PCR buffer II, 0.24 mm of each dNTP, and 0.24 mm 39- and 59-primer mixture of each specific outer pair and covered with 50 ml light white oil. PCR cycles were initiated by heading to 95 C for 5 min to denature all proteins and DNA; then at 95 C, 2.5 U AmpliTaq DNA polymerase (Perkin Elmer) were added to a total volume of 50 ml. PCR was carried out for 30 cycles of 45 sec at 94 C, 45 sec at 56 C, and 60 sec at 72 C. The reaction was terminated at 72 C for 6 min and quenched at 4 C. First round PCR products were stored at 220 C until the second round of PCR. For the second round PCR, 5 ml of the initial PCR products were added to the second PCR Mastermix to a total volume of 99.5 ml containing 1.9 mm MgCl2, 10 3 PCR buffer II, 0.2 mm of each dNTP, and 0.2 mm of a 39- and 59-primer mixture of each corresponding inner pair and covered with 50 ml light white oil. After completing the second round PCR (35 cycles) using the same program, samples were stored at 220 C until electrophoresis. Two percent agarose gel (Life Technologies) electrophoresis was carried out in an H5 electrophoresis chamber. Gels were stained with ethidium bromide (Sigma Chemical Co.). Twenty-five microliters of each PCR product and dye buffer were analyzed in parallel with a 100-bp DNA ladder (Life Technologies) as a standard. After completion of electrophoresis, the gel blot was analyzed, and photocopies of the blot were printed on the GelDoc 1000 system (Bio-Rad Laboratories, Inc., Hercules, CA).

TABLE 1. Outer and inner pairs of specific primers for mRNA amplification in RT-PCR mRNA

b-actin

Primer pair for PCR

Outer pair for first PCR Inner pair for second PCR

GnRH

Outer pair for first PCR Inner pair for second PCR

GnRH receptor

Outer pair for first PCR Inner pair for second PCR

Primers 59-39

Sequence of oligonucleotide

59-End 39-End 59-End 39-End

GTG CTC CAA CAG

GGC TTT GGT GAT

CGC GAT GTG GGC

TCT GTC ATG GTG

AGG ACG GTG AGG

CAC CAC GGA GAG

CAA GAT TTC ATG G AGC A

59-End 39-End 59-End 39-End

GCC TCT CAG CTC

GGC GCC CAC CTC

ATT TGG TGG GCA

CTA CTT TCC GAT

CTG CCT TAT CCC

CTG CTT GGG TGA

ACT CA TTG G

59-End 39-End 59-End 39-End

CAC GGT CAG CTG

TGG AGC TGG TAG

ATG GAA TAT TTT

GGA TGC GCT GCG

TGT GAC GGG TGG

GGA TGT GAG GTC

ATA CA TTC TTG

Product size (bp)

540 407 243 163 533 432

GnRH IN MURINE EMBRYONAL DEVELOPMENT

3707

TABLE 2. Distribution of embryos across embryonic stages for each treatment group with GnRH antagonist GnRH antagonist (mM)

0

0.05

0.1

0.5

1

2.5

5

10

2 cell (%) 8 cell (%) Morula (%) Blastocyst (%) Hatching blastocyst (%)

100 82 75 70 66

100 84 79 73 68

100 74 69 62 56

100 69 64 54 50

100 68 60 53 48

100 43 37 30 22

100 12 4 0 0

100 8 2 0 0

The identities of all PCR products after gel electrophoresis were confirmed by sequence analysis using the chain termination method (24, 25).

Immunohistochemical staining Single embryos from 88 h post-hCG (compacted morula; n 5 60), 95 h post-hCG (early expanded blastocyst; n 5 60), and 120 h post-hCG (hatching blastocyst; n 5 60) were examined by an avidin-biotin alkaline phosphatase technique (15) to localize at the protein level the GnRH in the preimplantation mouse embryo. Embryos were fixed with 4% paraformaldehyde in PBS for 30 min at 4 C in microdrops under oil and then treated with acid Tyrode’s solution (pH 2.5) to induce permeabilization of the zona pellucida. To reduce the nonspecific binding, 2% normal goat serum in PBS was applied to the embryos for 30 min at room temperature, then rinsed twice in PBS (pH 7.4) with 0.05% Tween-20 (PBS-T; Sigma Chemical Co.) and incubated for 90 min at 37 C with the primary antibody, polyclonal rabbit anti-[Lys8]GnRH, at 1 mg/ml (Sigma Chemical Co. and Petraglia). After being rinsed with PBS-T, embryos were incubated for 90 min at room temperature with a secondary antibody, biotinylated antirabbit IgG, at a dilution of 1:800 (Sigma Chemical Co.). Immunohistochemical controls were incubated with PBS containing 2% goat serum without primary antibody. To amplify the signal, embryos were washed with PBS-T, and then the avidin-biotin alkaline phosphatase-staining method (Vector Laboratories, Inc., Burlingame, CA) was used. Endogenous alkaline phosphatase activity was inhibited by the addition of levamisole to the buffer used to prepare the substrate solution. Finally, embryos were incubated in alkaline phosphatase substrate solution until color become evident, then the reaction was stopped in all the embryos simultaneously. A red precipitate indicated positive staining by the primary antibody. Embryos in microdrops under oil were visualized and photographed by a 35-mm camera (Olympus Corp.) attached to an inverted microscope (Olympus Corp.). The intensity of the immunostaining was evaluated by HSCORE (15). This method provides a numeric value of the overall staining intensity and the percentage of cells per embryo stained. HSCORE is calculated by the following equation: HSCORE 5 (Pi (i 1 1), where i is the intensity of staining with a value (obtained by two of the authors in a double blind manner) of 1 (weak), 2 (moderate), or 3 (strong), and Pi is the percentage of stained embryos in every stage for each intensity.

Embryo culture Exp 1. Two-cell embryos (n 5 785) were removed, washed with fresh medium, and then randomly transferred to 20-ml drops (an average of 15 embryos/drop) of modified Ham’s F-10 containing 0.3% BSA (Sigma Chemical Co.) (19) and 0, 0.05, 0.1, 0.5, 1, 2.5, 5, and 10 mm GnRH agonist (pGlu-His-Trp-Ser-Tyr-dHis(Bzl)-Leu-Arg-Pro-AzaGlyNH2, Histrelin, Sigma Chemical Co.) and 0, 0.05, 0.1, 0.5, 1, 2.5, 5, and 10 mm GnRH antagonist (NACd2Nal-d4Cphe-dTrp-Ser-Tyr-dHarg(Et2)-Leu-ArgPro-dAlaNH2. Detirelix, Sigma Chemical Co.). Embryos were checked daily using an inverted microscope (Olympus Corp.) for 3 consecutive days to evaluate the effect of GnRH agonist and antagonist on early development of murine embryos in comparison with growth in control medium. Exp 2. Two-cell mouse embryos (n 5 386) were cultured in medium containing 5 mm GnRH antagonist and increasing concentrations of GnRH agonist (0.1, 0.5, 1, 5, and 10 mm) for 72 h); as a control, three groups were cultured with medium only, medium with 5 mm GnRH antagonist, and medium with 10 mm GnRH agonist, respectively. Em-

FIG. 1. Agarose gel showing the products of nested PCR amplification for b-actin (A), GnRH (B), and GnRH (C) receptor mRNA in single embryos at morula (no. 2), early expanded blastocyst (no. 3), and hatching blastocyst (no. 4). Negative control samples of medium in which the embryos were cultured subjected to the same RT-PCR reaction are shown (no. 1).

bryos were checked daily using an inverted microscope (Olympus Corp.) to evaluate embryo development in each group. Both experiments were performed a minimum of three times with similar results.

Statistical analysis ANOVA and x2 test were employed to compare groups. Tukey’s and Scheffe’s tests were applied when ANOVA revealed statistical differences. The statistical analysis was carried out using the Statistical Package for Social Science (SPSS, Inc., Chicago, IL), and P , 0.05 was considered statistically significant.

Results Expression of mRNA transcripts of b-actin, GnRH, and GnRH receptor in individual mouse embryos at different developmental stages

A total of 180 embryos cultured with medium alone were examined by RT-PCR. Electrophoresis of RT-PCR fragments of each individual mouse embryo produced three different sized bands corresponding to fragment sizes produced by specific primers used in the second round PCR of DNA fragments for b-actin, GnRH, and GnRH receptor at embryo stages of compact morula, early expanded blastocyst, and hatching blastocyst, as shown in Fig. 1. All of the embryos at each developmental stages expressed b-actin mRNA, thus confirming the integrity of the RNA and the RT-PCR process. The percentage of embryos expressing GnRH receptor mRNA did not increase during in vitro development (morula to hatching blastocyst stage), remaining constant at 85–90%. The percentage of embryos positive for the expression of GnRH mRNA rose significantly (P , 0.05)

3708

GnRH IN MURINE EMBRYONAL DEVELOPMENT

Endo • 1999 Vol 140 • No 8

during embryo development from compacted morula (65%) to blastocyst (87%) and hatching blastocyst (90%) stages.

(P , 0.05) between the morula stage (2.83) and the blastocyst stage (early expanded, 3.83; hatching, 4.00).

Immunohistochemical study

Embryo culture

Immunoreactive GnRH was identified in all 180 mouse embryos studied (60 at compacted morula stage, 60 at early expanded blastocyst stage, and 60 at hatching blastocyst stage) during the preimplantation period using an avidinbiotin-alkaline phosphatase technique. To dispel any doubt about the specificity of the primary antibody used, two different antibodies against GnRH were used with the same results. Immunohistochemical localization of GnRH showed intense staining in the different blastomeres at morula stage as well as in the trophectoderm and inner cell mass of blastocysts (early expanded and hatching; Fig. 2). The comparison of immunostaining (HSCORE) assessed at each developmental stage using ANOVA with Scheffe’s analysis revealed statistical differences among different developmental stages (15). This method that provides a numeric value for the overall staining intensity and the percentage of embryos stained showed a significant increase

Exp 1. The percentage of two cell mouse embryos reaching the hatching blastocyst stage was significantly (P . 0.05) enhanced by culture with increasing concentrations of GnRH agonist (Fig. 3A) and was significantly decreased (P , 0.01) by culture with increasing concentrations of GnRH antagonist (Fig. 3B) compared with that in the control group cultured with medium only. Embryonic arrest occurs mostly between the two-cell and eight-cell stages. Moreover, GnRH antagonist (5 and 10 mm) was able to completely block embryo development, so no embryo reached the blastocyst stage at these concentrations. Exp 2. To illustrate that the detrimental effect of the GnRH antagonist on preimplantation embryonic development was a specific receptor-mediated effect, rather than a toxic effect, a second experiment was set up. In this second experiment, the deleterious effect of GnRH antagonist (5 mm) on embryo development was totally reversed in a dose-dependent man-

FIG. 2. Immunohistochemical localization of GnRH in morula (B) and hatching blastocyst (D). No staining was detected in negative controls, from which primary antibody was omitted, in morula (A) and hatching blastocyst (C). B, Intense GnRH staining can be localized in the different blastomeres of morula. D, Blastocyst stained positive for GnRH with increased immunostaining present in trophectoderm and inner cell mass. Original magnification, 3400.

GnRH IN MURINE EMBRYONAL DEVELOPMENT

FIG. 3. Percentage of two-cell mouse embryos that reached the hatching blastocyst stage at 95 h post-hCG after culture with increasing concentrations of GnRH agonist and antagonist. A, Two-cell mouse embryos were cultured in medium with 0 (n 5 100), 0.05 mM (n 5 45), 0.1 mM (n 5 45), 0.5 mM (n 5 45), 1 mM (n 5 45), 2.5 mM (n 5 47), 5 mM (n 5 46), and 10 mM (n 5 52) GnRH agonist. B, Two-cell mouse embryos were cultured in medium with 0 (n 5 100), 0.05 mM (n 5 48), 0.1 mM (n 5 49), 0.5 mM (n 5 46), 1 mM (n 5 51), 2.5 mM (n 5 49), 5 mM (n 5 54), and 10 mM (n 5 53) GnRH antagonist. Asterisks denote a statistical significant difference (P . 0.05) from control groups.

ner by increasing concentrations of the agonist, as determined by the number of embryos that reached the eight-cell (68 h post-hCG), compacted morula (88 h post-hCG), and blastocyst stage (95 h post-hCG) compared with values in the control group with medium only. The hatching rate (120 h post-hCG), however, was only partially reversed compared with the blastocyst rate (Fig. 4, A and B). Discussion

By the isolation, determination of structure, and synthesis of GnRH, more than 3000 agonistic or antagonistic analogues

3709

of this hormone have been synthesized (26). The use of GnRH agonist in assisted reproductive techniques has been generally accepted as the best way to induce multiple follicular development in combination with exogenous gonadotropins. The use of these GnRH analogs in in vitro fertilization has been associated with increased number of mature oocytes, rate of fertilization, and overall cycle fecundity rate (27). Although the principal function of GnRH is to stimulate the pituitary gland to release LH and FSH, there is evidence that an extrahypothalamic GnRH might have an important role in the reproductive process (2). A role for GnRH in modulating ovarian function has been described, involving steroidogenesis and oocyte maturation (28 –30). GnRH and GnRH agonist induce resumption of meiosis in follicle-enclosed oocytes both in vivo (31) and in vitro (32). This effect is mediated through a specific receptor (33) and involves the activation effect of GnRH agonist and antagonist on preimplantation embryonic development. Therefore, our first objective was to show that both GnRH and its receptor are expressed at the mRNA level in vitro by cultured mouse embryos during the preimplantation development period (morula to hatching blastocyst stages). Moreover, we were also able to demonstrate the presence of an immunoreactive GnRH in the different preimplantation embryos studied (morula to hatching blastocyst stages). Our data from both RT-PCR and immunohistochemical examination demonstrate that this hormone is produced as early as the morula stage. This observation is similar to that previously reported in rhesus monkey embryos during the entire periattachment period (35). The amount of mRNA in preimplantation embryos is not accurately known. Moreover, it has been recently show that the different blastomeres of the same embryo express different amount of mRNA for b-actin and interleukin-1 receptor (36). This agrees with previous immunohistochemical studies in which each blastomere of morula as well as embryos in the same stage of development stain with different intensities (15). Thus, the fact that we were able to detect GnRH and GnRH receptor mRNA expression in 65–90% and 85–90%, respectively, of single embryos studied is consistent with the findings of previous studies using this method (14, 20). Immunohistochemical examination of preimplantation mouse embryos indicates that GnRH was localized in both the inner cell mass and the trophectoderm at the blastocyst stage. This is also consistent with previous reports of the presence of immunoreactive GnRH in the cytotrophoblast of prehatched blastocyst (35) and in placental cytotrophoblast (37). In addition, we localized this hormone in the different blastomeres at the compacted morula stage. It is remarkable that both GnRH mRNA and protein expression are increased in the hatching blastocyst stage compared with the morula stage, as this hormone has been recently implied as a possible important paracrine factor in the process of embryonic implantation (6, 7). On the other hand, the GnRH receptor was ascertained at a constant level (80 – 85%) in all of the developmental embryonic stages studied, reinforcing the hypothesis that the embryo communicates with the maternal tubal epithelium and endometrium

3710

GnRH IN MURINE EMBRYONAL DEVELOPMENT

Endo • 1999 Vol 140 • No 8

FIG. 4. Percentages of two-cell mouse embryos reaching the eight-cell and compacted morula stages at 67 and 88 h post-hCG (A) and the blastocyst and hatching blastocyst stages at 95 and 120 h post-hCG (B) after culture in medium with GnRH antagonist (5 mM) and increasing concentrations of GnRH agonist: 0.1 mM (n 5 46), 0.5 mM (n 5 47), 1 mM (n 5 49), 5 mM (n 5 49), and 10 mM (n 5 51). Negative controls with medium only (n 5 45), medium with 10 mM GnRH agonist (n 5 52), and medium with 5 mM GnRH antagonist (n 5 47) were also established. Asterisks denote a statistical significant difference (P . 0.05) from other groups.

through the GnRH system to promote embryonic development and endometrial receptivity (10 –13). Trophoblastic GnRH has been implicated as one of the primary regulators of the synthesis and secretion of hCG both in periimplantation embryos (35) and in the placenta (5). Furthermore, the role of the inner cell mass in the induction of hCG synthesis and secretion by the trophoblast of the periimplantation primate blastocyst has previously been suggested (38). The intimate cell contact between the inner cell mass and the trophoblast seems to be necessary to initiate hCG synthesis (38). This along with evidence that GnRH secretion precedes hCG secretion in periimplantation embryos (35) may explain the fact that we localized the presence of GnRH not only in the trophectoderm, but also in the inner cell mass of the mouse blastocyst. This reinforces the poten-

tially important role of GnRH in controlling hCG synthesis and secretion not only in the placenta but also in the periimplantation embryos. The results from our in vitro culture of preimplantation mouse embryos exposed to GnRH agonist and antagonist suggest that GnRH is a crucial key in early embryonic development. The GnRH agonist seems to enhance embryonic development, whereas the GnRH antagonist has a detrimental effect. Further, GnRH antagonist is able to completely block early embryonic development, and the reversal of this effect by the agonist in a dose-dependent fashion suggests a specific receptor-mediated effect, rather than a nonspecific or toxic effect. This is consonant with the fact that exogenous administration of GnRH can displace the GnRH antagonist from the pituitary receptors and reestablish gonadotropin

GnRH IN MURINE EMBRYONAL DEVELOPMENT

secretion, as both GnRH and GnRH antagonist compete for binding with the same receptor (39). Moreover, there is evidence that both GnRH and GnRH agonist significantly increased the cleavage rate of bovine oocytes fertilized in vitro as well as that this effect was abolished by the addition of a GnRH antagonist (40). The direct action of both GnRH agonist and antagonist in vitro on preimplantation murine embryonic development is consistent with previous in vivo experiments in which GnRH agonist-treated mice had a higher rate of embryos reaching the hatching blastocyst stages as well as a higher pregnancy and implantation rate (30). Moreover, we have recently reported that infertile woman undergoing in vitro fertilization and embryo transfer had a significantly higher pregnancy and implantation rate if the administration of GnRH agonist was maintained during the early stages of embryonic development and implantation, suggesting a direct effect of the agonist on the embryo and maternal endometrium (41). Mammalian embryos must emerge, or hatch from their extracellular coat, the zona pellucida, to implant in the uterus at the blastocyst stage of development (42). The exact mechanism of hatching remains controversial. However, previous reports have related PG, trypsin-like substance, plasminogen and plasmin, glutamine, and such phenomena as fluid accumulation and expansion to the mechanism of hatching (42, 43). PGs have been implicated as remarkable participants in the hatching process during the development of a variety of animal species (43). It is interesting that the detrimental effect of GnRH antagonist on preimplantation embryonic development can be completely reversed by the agonist with development to the blastocyst stage. However, the hatching rate was only partially reversed. This could be related to the fact that GnRH is modulating the production and release of PGE2 and PGF2a in the trophoblast (44), so the GnRH antagonist may be interfering the hatching process by affecting PG production. Furthermore, it has been shown that coculture of mouse blastocysts with human placental cells, which are known to produce GnRH, enhances the hatching rate (45). This is also consistent with previous reports that embryos failing to hatch in vitro secreted very low amounts of GnRH compared with those that did hatch and attach (35). It has also been previously demonstrated in mice that the administration of GnRH agonist in vivo significantly increases germinal vesicle breakdown and in vitro fertilization rates of these oocytes. Additionally, these fertilized oocytes developed more rapidly than the control group (30). This is consistent with our in vitro findings that embryos cultured with increasing concentrations of GnRH agonist displayed enhanced preimplantation development. On the basis of the observations just described it is tempting to suggest that GnRH may play an important role in preimplantation mammalian embryo development, endometrial receptivity, and implantation. Acknowledgments We are grateful to Prof. Felice Petraglia, University of Udine (Udine, Italy), for providing us with the GnRH antibody.

3711

References 1. Conn PM, Crowley Jr WF 1991 Gonadotropin releasing hormone and its analogues. N Engl J Med 324:93–103 2. Hsueh AJW, Jones PBC 1981 Extrapituitary actions of gonadotropin releasing hormone. Endocr Rev 2:437– 455 3. Chieffi G, Pierantoni R, Fasano S 1991 Immunoreactive GnRH in hypothalamic and extrahypothalamic areas. Int Rev Cytol 127:1–55 4. Imai A, Furui T, Tamaya T 1992 Is extrapituitary action of gonadotropin releasing hormone biologically significant? Ann Clin Biochem 29:477– 480 5. Prager D, Weber MM, Herman-Bonert V 1992 Placental growth factors and releasing/inhibiting peptides. Semin Reprod Endocrinol 10:83–91 6. Casan˜ EM, Raga F, Kruessel JS, Wen Y, Nezhat C, Polan ML 1998 Immunoreactive gonadotropin-releasing hormone expression in cycling human endometrium of fertil patients. Fertil Steril 70:102–106 7. Raga F, Casan˜ EM, Kruessel JS, Wen Y, Huang HY, Nezhat C, Polan ML 1998 Quantitative gonadotropin-releasing hormone (GnRH) gene expression and immunohistochemical localization in human endometrium throughout the menstrual cycle. Biol Reprod 59:661– 669 8. Emons G, Schroder B, Ortmann O, Westphalen S, Schulz K, Schally AV 1993 High affinity binding and direct antiproliferative effects of luteinizing hormone releasing hormone analogs in human endometrial cancer lines. J Clin Endocrinol Metab 77:1458 –1464 9. Paria BC, Huet-Hudson YM, Dey SK 1993 Blastocyst’s state of activity determines the “window” of implantation in the receptive mouse uterus. Proc Natl Acad Sci USA 90:10159 –10162 10. Tazuke SI, Guidice LC 1996 Growth factors and cytokines in endometrium, embryonic development, and maternal: embryonic interactions. Semin Reprod Endocrinol 14:231–243 11. Das SK, Wang X-N, Paria BC, Damm D, Abraham JA, Klagsbrun M, Andrews GK, Dey SK 1994 Heparin-binding EGF-like growth factor gene is induced in the mouse uterus temporally by the blastocyst solely at the site of its apposition: a possible ligand for interaction with blastocyst EGF-receptor in implantation. Development 120:1071–1083 12. Hearn JP, Gidley-Baird AA, Hodges JK, Summers PM, Webley GE 1988 Embryonic signals during the peri-implantation period in primates. J Reprod Fertil 36:49 –58 13. Asirvatham AL, Johnson GA, Belden EL, Van Kirk EA, Moss GE, Murdoch WJ 1994 Immunization of mice against a synthetic N-terminal extracellular domain gonadotropin-releasing hormone receptor peptide: evidence for a direct uterine effect. Am J Reprod Immunol 32:95–100 14. Huang HY, Kruessel JS, Wen Y, Polan ML 1997 Use of reverse transcriptionpolymerase chain reaction to detect embryonic interleukin-1 system messenger RNA in individual preimplantation mouse embryos co-cultured with Vero cells. Hum Reprod 12:1537–1544 15. De los Santos MJ, Mercader A, Frances A, Portoles E, Remohi J, Pellicer A, Simon C 1996 Role of endometrial factors in regulating secretion of components of the immunoreactive human embryonic interleukin-1 system during embryonic development. Biol Reprod 54:563–574 16. Murdoch WJ 1995 Immunolocalization of a gonadotropin-releasing hormone receptor site in murine endometrium that mediates apoptosis. Cell Tissue Res 282:527–529 17. Balasch J, Martinez F, Jove´ I., Cabre´ L, Coroleu B, Barri PN, Vanrell JA 1993 Inadvertent gonadotrophin-releasing hormone agonist (GnRHa) administration in the luteal phase may improve fecundity in in-vitro fertilization patients. Hum Reprod 8:1148 –1151 18. Gartner B, Moreno C, Marinaro A, Remohi J, Simon C, Pellicer A 1997 Accidental exposure to daily long-acting gonadotrophin-releasing hormone analogue administration and pregnancy in an in-vitro fertilization cycle. Hum Reprod 2:2557–2559 19. Barroso PB, Osuamkpe C, Nagamani M, Yallampalli C 1998 Nitric oxide inhibits development of embryos and implantation in mice. Mol Hum Reprod 4:503–507 20. Kruessel JS, Huang HY, Wen Y, Kloodt AR, Bielfeld P, Polan ML 1997 Different pattern of interleukin-1b, interleukin-1 receptor antagonist and interleukin-1 receptor type I mRNA-expression in single preimplantation mouse embryos at various developmental stages. J Reprod Immunol 34:103–120 21. Alonso S, Minty A, Bourlet Y, Buckingham ME 1986 Comparison of three actin-coding sequences in the mouse: evolutionary relationships between the actin genes of warm-blooded vertebrates. J Mol Evol 23:11–22 22. Mason AJ, Hayflick JS, Zoeller RT, Young WS, Phillis HS, Nikolics K, Seeburg PH 1986 A deletion truncating the gonadotropin-releasing hormone gene is responsible for hypogonadism in the hpg mouse. Science 234:1366 –1371 23. Tsutsumi M, Zhou W, Millar RP, Mellon PL, Roberts JL, Flanagan CA, Dong K, Gillo B, Sealfon SC 1992 Cloning and functional expression of a mouse gonadotropin-releasing hormone receptor. Mol Endocrinol 6:1163–1169 24. Sanger F, Coulson AR 1975 A rapid method for determining sequences in DNA by primed synthesis with DNA polymerase. J Mol Biol 94:441– 448 25. Sanger F, Nicklen S, Coulson AR 1992 DNA sequencing with chain-terminating inhibitors. Biotechnology 24:104 –108 26. Emons G, Schally AV 1994 The use of luteinizing hormone releasing hormone

3712

27. 28. 29.

30. 31. 32. 33. 34. 35.

36.

GnRH IN MURINE EMBRYONAL DEVELOPMENT

agonist and antagonist in gynecological cancers. Hum Reprod Update 9:1364 –1379 Testart J, Lefevre B, Gougeon A 1993 Effects of gonadotrophin-releasing hormone agonist (GnRHa) on follicle and oocyte quality. Hum Reprod 8:511–518 Knecht M, Ranta T, Feng P, Shinohara O, Catt KJ 1985 Gonadotropin releasing hormone as a modulator of ovarian function. J Steroid Biochem 23:771–778 Pellicer A, Tarin JJ, Miro F, Sampaio M, De los Santos MJ, Remohi J 1992 The use of gonadotrophin releasing analogues (GnRHa), in in-vitro fertilization: some clinical and experimental investigations of a direct effect on the human ovary. Hum Reprod 7:39 – 47 Yang BC, Uemura T, Minaguchi H 1995 Effects of a gonadotropin releasing hormone agonist on oocyte maturation, fertilization, and embryonal development in mice. J Assit Reprod Genet 12:728 –732 Banka CL, Erickson GF 1985 Gonadotropin-releasing hormone induces classical meiotic maturation in subpopulations of atretic preantral follicles. Endocrinology 117:1500 –1507 Hillensjo¨ T, LeMaire WJ 1980 Gonadotropin-releasing hormone agonists stimulate meiotic maturation of follicle-enclosed rat oocytes. Nature 287:145–146 Dekel N, Lewysohn O, Ayalon D, Hazum E 1988 Receptors for gonadotropinreleasing hormone are present in rat oocytes. Endocrinology 123:1205–1207 Aberdam E, Dekel N 1985 Activators of protein kinase C stimulate meiotic maturation of rat oocytes. Biochem Res Commun 132:570 –574 Seshagiri PB, Terasawa E, Hearn JP 1994 The secretion of gonadotropinreleasing hormone by peri-implantation embryos of the rhesus monkey: comparison with the secretion of chorionic gonadotropin. Hum Reprod 9:1300 –1307 Kruessel JS, Huang HY, Simon C, Behr B, Pape AR, Wen Y, Bielfeld P, Polan

37. 38. 39.

40. 41.

42. 43. 44.

45.

Endo • 1999 Vol 140 • No 8

ML 1998 Single blastomeres within human preimplantation embryos express different amounts of messenger ribonucleic acid for b-actin and interleukin-1 receptor type I. J Clin Endocrinol Metab 83:953–959 Khord GS, Siler-Khord TM 1978 Localization of luteinizing hormone-releasing hormone in the human placenta. Fertil Steril 29:523–526 Summers PM, Taylor CT, Miller MW 1993 Requirement of inner cell mass for efficient chorionic gonadotrophin secretion by blastocyst of common marmosets (Callithrix jacchus). J Reprod Fertil 97:321–327 Gordon K, Williams RF, Danforth DT, Hodgen GD 1990 A novel regimen of gonadotropin-releasing hormone (GnRH) antagonist plus pulsatile GnRH: controlled restoration of gonadotropin secretion and ovulation induction. Fertil Steril 54:1140 –5 Funston RN, Seidel GE 1995 Gonadotropin-releasing hormone increases cleavage rates of bovine oocytes fertilized in vitro. Biol Reprod 53:541–545 Raga F, Casan˜ EM, Ruiz A, Remohi J, Simon C, Polan ML, Pellicer A 1998 Gonadotropin-releasing hormone agonist (GnRH-a) administered throughout the luteal phase and early pregnancy increases fecundity in IVF-ET patients. Hum Reprod 13:93 Perona RM, Wassarman PM 1986 Mouse blastocyst hatch in vitro by using a trypsin-like proteinase associated with cells of mural trophectoderm. Dev Biol 114:42–52 Bigger JD, Leonov BV, Baskar JF, Fried J 1978 Inhibition of hatching of mouse blastocyst in vitro by prostaglandin antagonist. Biol Reprod 19:519 –533 Petraglia F, Lim AT, Vale W 1987 Adenosine 39,59-monophosphate, prostaglandins, and epinephrine stimulate the secretion of immunoreactive gonadotrophin-releasing hormone from cultured human placental cells. J Clin Endocrinol Metab 65:1020 –1025 Morishita T, Yokoyama M, Nozaki M, Sano M, Nakano H 1993 Improvement in blastocyst hatching of mouse embryos cocultured with human placental cells. J Assist Reprod Gen 10:463– 467