Skip to main content
Journal of Assisted Reproduction and Genetics logoLink to Journal of Assisted Reproduction and Genetics
. 2016 May 26;33(8):1027–1039. doi: 10.1007/s10815-016-0735-z

Commonly used fertility drugs, a diet supplement, and stress force AMPK-dependent block of stemness and development in cultured mammalian embryos

Alan Bolnick 1,, Mohammed Abdulhasan 1, Brian Kilburn 1, Yufen Xie 2, Mindie Howard 3, Paul Andresen 4, Alexandra M Shamir 5, Jing Dai 1, Elizabeth E Puscheck 1, Daniel A Rappolee 1,6,7,8,9
PMCID: PMC4974229  PMID: 27230877

Abstract

Purpose

The purpose of the present study is to test whether metformin, aspirin, or diet supplement (DS) BioResponse-3,3′-Diindolylmethane (BR-DIM) can induce AMP-activated protein kinase (AMPK)-dependent potency loss in cultured embryos and whether metformin (Met) + Aspirin (Asa) or BR-DIM causes an AMPK-dependent decrease in embryonic development.

Methods

The methods used were as follows: culture post-thaw mouse zygotes to the two-cell embryo stage and test effects after 1-h AMPK agonists’ (e.g., Met, Asa, BR-DIM, control hyperosmotic stress) exposure on AMPK-dependent loss of Oct4 and/or Rex1 nuclear potency factors, confirm AMPK dependence by reversing potency loss in two-cell-stage embryos with AMPK inhibitor compound C (CC), test whether Met + Asa (i.e., co-added) or DS BR-DIM decreases development of two-cell to blastocyst stage in an AMPK-dependent (CC-sensitive) manner, and evaluate the level of Rex1 and Oct4 nuclear fluorescence in two-cell-stage embryos and rate of two-cell-stage embryo development to blastocysts.

Result(s)

Met, Asa, BR-DIM, or hyperosmotic sorbitol stress induces rapid ~50–85 % Rex1 and/or Oct4 protein loss in two-cell embryos. This loss is ~60–90 % reversible by co-culture with AMPK inhibitor CC. Embryo development from two-cell to blastocyst stage is decreased in culture with either Met + Asa or BR-DIM, and this is either >90 or ~60 % reversible with CC, respectively.

Conclusion

These experimental designs here showed that Met-, Asa-, BR-DIM-, or sorbitol stress-induced rapid potency loss in two-cell embryos is AMPK dependent as suggested by inhibition of Rex1 and/or Oct4 protein loss with an AMPK inhibitor. The DS BR-DIM or fertility drugs (e.g., Met + Asa) that are used to enhance maternal metabolism to support fertility can also chronically slow embryo growth and block development in an AMPK-dependent manner.

Keywords: Embryo culture, Fertility drugs, Diet supplement, Embryo development, Embryo quality

Introduction

Drugs such as metformin (Met) or diet supplements (DSs) such as BioResponse-3,3′-Diindolylmethane (BR-DIM) are used for improving fertility and diabetic metabolism. Aspirin (Asa) is regularly utilized for analgesia and as an anti-inflammatory pharmaceutical. Met is commonly used to support women with ovulatory problems due to polycystic ovarian syndrome (PCOS) [13] and may be used by women with type 2 diabetes (T2D), and aspirin (Asa) is used by fertile and infertile women episodically for pain relief [2, 413] and recurrent pregnancy loss due to anti-phospholipid syndrome [14]. Met [15] and Asa [16] may have part of their therapeutic activity through the activation of AMP-activated protein kinase (AMPK). BR-DIM is a commonly used diet extract of yellow cruciferous vegetables [17]. BR-DIM is used to treat women with reproductive problems such as cervical dysplasia [18] and can affect pregnancy [19]. Consumption of cruciferous vegetables is associated with lower prostate cancer risk [20], and cruciferous diet extract BR-DIM slows growth and causes human prostate cancer cell death, in vitro and in vivo in an AMPK-dependent manner [21]. However, BR-DIM slows growth or kills cancer stem cells, but in embryos, this effect on stem cells would be detrimental. Thus, diet supplements (DSs) like BR-DIM, and drugs such as Asa and Met, may have therapeutic effects through AMPK activity, but we hypothesize that high-dose ranges may be toxic for early embryos.

AMPK helps oocyte or blastocyst metabolism that is already compromised and helps IVM that has high levels of stress and has high oocyte or embryo loss rate due to diabetes [2225]. Met or other AMPK agonists are often studied for their positive effects on maternal metabolism or when pathogenic insults are present. In vivo AMPK can reverse 88 % resorption rate caused by dehydroepiandrosterone (DHEA); however, rescue to 45 % resorption does not match 35 % normal resorption rate [26]. Similarly, clomiphene and Met reverse low fecundity of PCOS patients, but there is loss of chemical pregnancy in Met metformin alone compared with clomiphene and metformin [12]. However, in most studies, no metformin-alone control is done to test for possible toxicity in oocytes and embryos not under severe stress. We hypothesize that many AMPK activators occur in diets and drugs, and oocytes and embryos that are not under pathogenic stimuli may have a toxic AMPK activation levels that are too high [22, 23, 27, 28].

Hyperosmotic stress led to the cloning of the first stress enzyme in the yeast model for sporulation forced by starvation [29]. The cloning of the first stress enzymes in mammals also used hyperosmotic stress, and this has been used as the positive control by many labs that study stress enzymes in somatic cells or study stress or stress enzymes in reproductive systems [3032]. Use of hyperosmotic stress enables comparison of results between stem cells and embryos and is universal stress for all cells.

The AMPK heterotrimer has a catalytic α-subunit, positive regulatory γ-subunit, and negative regulatory β-subunit [33, 34]. Met activates the positive regulatory γ-subunit through increasing AMP and decreasing adenosine triphosphate (ATP) [35] or in an ATP-independent manner [36], and Asa decreases the activity of the negative regulatory β-subunit by binding ser108 [16]. Thus, Met [35] and Asa [16] activate the AMPK heterotrimer in indirect and direct mechanisms through different subunits, and therapeutic mechanisms occur through AMPK. But, many DSs or drugs may have additive effects that supersede the beneficial dose range into the toxic range. Many DSs and drugs may have supramaximal effects in regulation of cancer cells and an impact in the liver [3739]. Additive effects could be from simultaneous exposure or sequential exposure due to dietary or drug consumption.

AMPK is beneficial to mouse oocytes under stress [22, 27, 28, 40] and oocytes and embryos isolated from diabetic mothers and subject to diabetic hyperglycemia and dysfunctional insulin signaling [24, 25, 28]. But, AMPK activation occurring in normal preimplantation blastocysts and cultured embryonic stem cells (ESCs) and trophoblast stem cells (TSCs), isolated from the blastocyst [41], leads to loss of potency factors and inhibitor of differentiation (Id)2, caudal domain homeobox (Cdx)2 in blastocysts, octamer-binding transcription factor (Oct)4 and nanog in pluripotent ESCs and inducible pluripotent stem cells (iPSCs) [42, 43], and Cdx2 and Id2 in TSCs [41, 4446] and in two-cell-stage embryos [41, 45]. Oct4 is necessary for the ESC stress response and survival [4749] and is necessary for metabolic control in the blastocyst [50], and null Oct4 mutants are lethal at the blastocyst stage due to insufficient function of cells of the inner cell mass (ICM)/ESC lineage cells [51]. Of significance is that small variations in Oct4 level, including loss in the levels caused by stress, change the fate of the ESC [52]. Rex1 is expressed in the ICM also and, although not necessary for embryo survival, is lost when ICM cells follow either of its immediate cell fates, extraembryonic primitive endoderm, or primitive ectoderm that produces all tissues at gastrulation [30]. Thus, it is important to move from potency factor loss of TSC potency factors to testing for the loss of ESC potency factors that will mark and control the pluripotency of stem cells in the embryo that ultimately produce the neonate.

However, these previous studies used hyperosmotic, hypoxic, and genotoxic stressor but did not study DSs or drugs that are known to activate AMPK. If potency loss is associated with negative effects, AMPK-activating drugs and DSs may cause toxic effects in embryos. Thus, we hypothesize that AMPK-activating drugs, such as Met and Asa, and DSs, such as BR-DIM, can cause potency loss in two-cell embryos and lead to decreased developmental rates in cultured mouse embryos. This hypothesis is tested here.

Materials and methods

Materials

Sorbitol, Asa (tissue culture-grade acetylsalicylic acid), and Met were from Sigma Chemical Co. (St. Louis, MO). The primary antibodies for total mouse monoclonal anti-Oct4 and rabbit polyclonal Rex1 were from Santa Cruz Biotechnology (Santa Cruz, CA). The AMPK inhibitor compound C (CC) was from Calbiochem (San Diego, CA). BR-DIM was from Dr. Dou, Wayne State University School of Medicine, and was prepared and used similar to protocols for in vitro treatment of human prostate cancer cells [21]. BR-DIM was purchased from BioResponse (BioResponse, Boulder, CO)

Embryo culture and treatment

Commercially available cryopreserved mouse zygotes from superovulated female B6C3F-1 × male B6D2F-1 mice (Embryotech Laboratories, Inc., Haverhill, MA, USA) were used. Both the test and control embryos were set up in triplicate under oil and cultured in 5 % CO2 at 37 °C until they were scored for development to expanded blastocysts. The one-cell mouse embryo assay noted and recorded the development from one-cell to two-cell in 24 h and one-cell to expanded blastocyst in 96 h. The two-cell mouse embryo assay only noted/recorded development from two-cell to expanded blastocyst in 72 h. Embryotech Laboratories Inc. (ELI) requires greater than 70 % blastocyst formation from the control group to validate the one-cell assay. Minimum blastocyst rate is 80 % for the two-cell assay. The quality of cryopreserved zygotes used in this study was validated by a very high blastocyst formation rate >90 % in vehicle, potassium Simplex optimized media (KSOM) with amino acids (KSOMAA; Global medium) (Fig. 2a) after 4 days of culture. Standard techniques were used for obtaining mouse embryos [53]. Female B6C3F1 mice (3–4 weeks old, Envigo, Indianapolis, IN) were super-ovulated and mated with male B6D2F1 mice. After superovulation and mating, the female B6C3F1 mice were euthanized and the oviducts containing the one-cell mouse embryos were harvested. The cumulus intact one-cell mouse embryos were removed from the oviduct and placed in hyaluronidase to remove all cumulus cells. The cumulus-free one-cell mouse embryos were rinsed in M2 medium with HEPES (Sigma® Life Science, Catalog M7167) before being placed into cryoprotectant (ethylene glycol-based cryopreservation medium) and loaded into 0.25-cc straws. Thawing was performed according to the manufacturer’s protocol. After thawing, the embryos were incubated at 37 °C and 5 % CO2 in KSOMAA for 18 h and examined for development. Embryos showing signs of fragmentation and delayed or accelerated development were discarded. In all studies, embryos were equilibrated for at least 1 h in lowest-stress KSOMAA [54] and stressed with the stimulus dose for the time period indicated. KSOMAA was 260–270 mOsmol, increasing 1.7-fold to 498 mOsmol with the addition of 200 mM sorbitol. For inhibitor studies (except where indicated), the inhibitors were preloaded with embryos 3 h before the stress and continued during the stress. In the text, the level of sorbitol (w/v) added is used to produce the given molarity of sorbitol [55, 56]. For inhibitor studies, the inhibitors were incubated with embryos for 2 h before stress was added and during stress. The dose of AMPK inhibitor CC used in this study was 5 μM. Embryos were preloaded with CC for 2 h to saturate endogenous AMPK prior to experimental agonists as done previously [57], and then, the embryos were treated with 200 mM sorbitol or 1 mM Met and/or 10 μM acetylsalicylic acid or 20 μM BR-DIM in the continuing presence of 5 μM CC for 1 h or for 3 days.

Fig. 2.

Fig. 2

AMPK antagonist compound C (CC) reverses retarded embryo development caused by AMPK agonist drugs (Met + Asa) and diet supplement (BR-DIM). Embryos were cultured overnight from day 0 to day 1, stimulated −/+ CC day 1 at the two-cell stage, and micrographed daily to assay effects on embryo development. From top to bottom, development was assayed after stimulation by a KSOMAA alone, b KSOMAA + CC, c Met + Asa, d Met + Asa + CC, e BR-DIM, and f BR-DIM + CC. On days 2, 3, and 4 embryos were categorized for development from the two-cell stage, compaction at eight-cell stage through morula. On days 3 and 4, the embryos were also categorized as blastocysts. Biological experiments were done in triplicate, and quantitative immunofluorescence of nuclei was done using Simple PCI DN module and analyzed for significance using ANOVA and Tukey post hoc test. aShows that BR-DIM or Met + Asa is different than KSOMAA control for time- and developmental stage-matched embryos (p < 0.05). bShows that BR-DIM + CC or Met + Asa + CC is significantly different compared to time- and developmental stage-matched embryos BR-DIM or Met + Asa, respectively (p < 0.05)

Met was used at 1 mM because it caused little cellular morbidity for 3 days of mouse ESC culture but regulated expression of paired box 3 (Pax3) transcription factor [58, 59]. BR-DIM was used at 20 μM because this dose was used to slow growth or kill cultured prostate cancer cells [21], and a peak of ~7 μM BR-DIM was reported during in vivo mouse exposure [60]. Asa (salicylate) was used at 10 μM because this is a standard plasma level after enteric pill [61] and well within the 1–3-mM levels after high-dose Asa treatment of T2D [62], and the 2-mM dose was used to show that major therapeutic levels of Asa require AMPK activity [16]. CC at 5 μM was validated in dose-response testing in other previous studies from our lab for testing AMPK effects in two-cell embryos and blastocysts and recently optimized for TSCs [41, 45, 63].

Immunofluoresence

Two-cell-stage embryos were fixed, quenched, permeabilized, and stained for Oct4 and Rex1 and counterstained for 4′,6-diamidino-2-phenylindole (DAPI) by modifying previous protocols used in mouse ESCs and validated by immunoblot [64, 65]. Briefly, embryos were fixed in 2 % paraformaldehyde in phosphate-buffered saline (PBS; pH 7.4) for 30 min at room temperature, quenched in 0.5 M glycine, and rinsed three times in PBS containing 0.5 % bovine serum albumin (PBS/BSA). After paraformaldehyde fixation, two-cell embryos were permeabilized for 15 min with 0.1 % Triton X-100. Potency factors were dual-labeled simultaneously overnight at 4 °C using a mouse monoclonal antibody against Oct4 and a rabbit polyclonal antibody against Rex1, and both diluted 1:100 from a 200 μg/ml stock (Santa Cruz Biotechnology, Inc., Santa Cruz, CA). Binding of primary antibodies was visualized using a combination of fluorescein isothiocyanate (FITC)-conjugated affinity-purified donkey anti-mouse immunoglobulin (IgG) and Texas Red-conjugated affinity-purified donkey anti-rabbit IgG (Jackson Immunoresearch Laboratories, Inc., West Grove, PA) diluted 1:500 from a 1 mg/ml stock. Embryos were incubated for 60 min at room temperature in the secondary antibody mixture, and nuclei were counterstained with DAPI diluted 1:1000 from a 5 mg/ml stock and mounted on glass slides. Fluorescent antibody labeling was imaged using a Hamamatsu Orca cooled-chip digital camera and a Leica DM IRB microscope with filter sets for DAPI, FITC, and Texas Red. Embryos were imaged at an objective magnification of ×20 and an exposure time of 2.0 s. The FITC or Texas Red stain intensities were quantified using Simple PCI (Hamamatsu) imaging software and formatted using Microsoft Excel and Adobe Photoshop 7.0. Fluorescence intensities (grey levels) were determined for each antibody and nonimmune IgG (background) by circumscribing the nuclei. All micrographs were exposed using the same shutter speed, and all experiments were repeated at least three times.

Statistical analysis

All experiments were performed with at least three replications. Data on immunofluorescence were analyzed with one-way analysis of variance (ANOVA) followed by Tukey’s post hoc comparisons. Percentages of embryos reaching developmental milestones were arcsine transformed before analysis. A general linear model was used to examine the main effects of stimulation treatments, days, and stages and their two-way and three-way interactions. The interactions between treatments and days and between days and stages were significant, allowing the subsequent comparisons among treatments on a specific day or at a specific stage using Dunnett’s and/or Tukey’s post hoc tests. The cell numbers per embryo were analyzed using one-way ANOVA followed by Tukey’s post hoc test to examine the difference among stimulation treatments on day 2. Independent t test was used to compare the cell numbers per embryo between days 2 and 4 for Met + Asa or BR-DIM. All analyses were performed in Statistical Package for the Social Sciences (SPSS) version 23.0 (SPSS Inc., Chicago, IL).

Results

We previously showed that standard positive control hyperosmotic stress caused AMPK-dependent Cdx2 and Id2 protein loss in two-cell embryos where the function of loss is not known and in embryos and TSCs where forced loss leads to depletion of stem cells and differentiation to first lineage [41, 45, 46]. Here, we test the hypothesis that clinically relevant drugs and a DSs cause AMPK-dependent potency factor loss and embryo development. The rationale for doses used for sorbitol, Met, Asa, BR-DIM, and CC is analyzed in the “Embryo culture and treatment” section of the “Materials and methods” section. Culture of embryos in KSOMAA sustained nuclear expression of Oct4 and Rex1 (Fig. 1a), and 1-h treatment of two-cell-stage embryos with 1 mM Met or 200 mM sorbitol decreased Oct4 by 64 and 57 %, respectively, and Rex1 by 53 and 52 %, respectively. The hyperosmotic stress- or drug-induced loss of Oct4 and Rex1 was significant (p < 0.05) (Fig. 1b). CC pretreatment and co-treatment with sorbitol or Met reversed Rex1 loss by 92 or 97 %, respectively (p < 0.05). CC pretreatment and co-treatment with sorbitol or Met reversed Oct4 loss 78 % or 85 %, respectively, and the reversal of hyperosmotic stress- or drug-induced loss of Oct4 and Rex1 were significant (p < 0.05). Rex1 loss caused by metformin was reversed by CC to a level that was not significantly different than Rex1 expression in unstressed embryos (p = 0.26). Thus, either positive control hyperosmotic sorbitol or Met causes significant loss of two nuclear, potency factor proteins and is AMPK dependent as AMPK antagonist reverses loss to the point where embryos have nearly the potency of unstressed embryos.

Fig. 1.

Fig. 1

AMPK mediates hyperosmotic stress- and Met AMPK agonist-induced loss of nuclear Oct4 and Rex1 potency factors that is largely reversed by CC (a). Zygotes were cultured overnight in lowest-stress media, some two-cell embryos were then preloaded with 5 μM CC for 2 h, and at time 0, embryos were incubated with 200 mM hyperosmotic stress or 1 mM Met for 1 h. Embryos were fixed, quenched, permeabilized, and exposed to polyclonal anti-Rex1 or monoclonal anti-Oct4 antibodies and counterstained with anti-rabbit TxRed or anti-mouse FITC and Hoechst and then micrographed. Embryos were treated with KSOMAA alone (aka unstimulated; AC), 200 mM sorbitol (DF), sorbitol with CC (GI), 1 mM Met (JL), or 5 μM CC with 1 mM Met (MO). The micrographs represent Hoechst (A, D, G, J, M), Oct4 (B, E, H, K, N), and Rex1 (C, F, I, L, O) after 1 h of the indicated treatments after exposures of the same embryos. Biological experiments were done in triplicate, and quantitative immunofluorescence of nuclei was done using Simple PCI DN module and analyzed for significance using ANOVA and Tukey post hoc test (b). AMPK induces loss of nuclear potency factors through hyperosmotic stress or the AMPK agonist Met, and loss is reversed by AMPK antagonist CC. Biological experiments were done in triplicate, and quantitative immunofluorescence of nuclei was done using Simple PCI DN module and analyzed for significance using ANOVA and Tukey post hoc test. aShows a significant difference compared with KSOMAA control (p < 0.05). bShows a significant difference between sorbitol and sorbitol + CC or between Met and Met + CC and a significant difference compared with KSOMAA (p < 0.05). cShows significant difference compared with Met (p < 0.05) but no significant difference compared with KSOMAA

KSOMAA was previously shown to be a very low-stress medium; it enabled highest developmental rates, and >90 % of zygotes reached the blastocyst stage by day 4 (Fig. 2a). CC (5 μM) also enabled high growth and development that was not significantly different from KSOMAA alone on days 2 and 3. However, by day 4, 64 % of embryos had reached blastocyst stage. Both Met + Asa (1 mM + 10 μM respectively) and 20 μM BR-DIM significantly (p < 0.05) slowed embryo development on days 2 and 3, and by day 4, only 3 % of two-cell-stage embryo in the Met + Asa group and 0 % of two-cell-stage embryo on the BR-DIM group had reached the blastocyst stage. However, co-culture with CC significantly (p < 0.05) reversed the nearly absolute block of development of 65.6 and 42.3 % of embryos to the blastocyst stage by day 4 after treatment with Met + Asa and BR-DIM, respectively. Although CC had some effects on day 2 for both BR-DIM and Met + Asa, more embryos were in the least developed state. It was not until day 4 that a strong reversal and over half the embryos had progressed to more advance developmental stages. By day 4, CC significantly (p < 0.05) reversed Met + Asa from 22.3 to 88.4 % and BR-DIM 0 to 71 %, Taken together, the data suggest that AMPK agonism is associated with slower early development followed by high levels of arrested development by the blastocyst stage

The data suggest that there is an optimal level of AMPK activity that is between addition of antagonist and addition of two agonists. In order to determine the effects of levels of AMPK modulation on development of embryos cultured from two-cell to blastocyst, we compared the micrographs and the fractions of embryos developed to blastocyst in increasing levels of AMPK modulation; single antagonist, no modulation, two agonists with one antagonist, one agonist, and two agonists. The data show that embryos with antagonist CC develop to blastocyst stage (64.6 ± 17.6 %) (Fig. 3) but are not of apparent high quality and would not typically be reimplanted in an IVF clinic. A higher rate of untreated embryos develops to blastocyst (90 ± 5.9 %) than embryos treated with two agonists and CC (65.6 ± 8.8 % blastocysts), and the untreated embryos have a larger apparent ICM and would be judged the best for reimplantation. Single agonists (BR-DIM) or double agonists (Met + Asa) produced less than 4 % blastocysts. Clearly, the reversal by CC of poor development caused by Met + Asa is apparent morphologically as the CC-reversed group is of generally high quality. Although, all stimulants have off-target effects; taken together, the data suggest an optimization of embryonic development of AMPK levels whereby medium alone is optimal and antagonism or agonism is suboptimal.

Fig. 3.

Fig. 3

Regulation of AMPK activity by AMPK agonists and antagonists is critical for embryo development and suggests an optimal AMPK activity (a). All zygotes were cultured overnight in lowest-stress media KSOMAA to adapt to culture. Prior to treatment (time 0), two-cell embryos were pretreated with vehicle or 5 μM CC for 2 h followed by treatment with vehicle, or AMPK agonists, 10 μM Asa, and 1 mM Met or with 20 μM BR-DIM alone until day 4 when final development was assayed from micrographs (one experiment shown above). All experiments were done in triplicate, and embryo development was assessed twice daily. Micrographs were taken and number of blastocysts was formed from initial two-cell-stage embryo recorded. Micrograph images show treatment with AMPK antagonist CC and AMPK antagonists Met + Asa, or BR-DIM has putative effects on embryo development (b). Biological experiments were done in triplicate, and quantitative immunofluorescence of nuclei was done using Simple PCI DN module and analyzed for significance using ANOVA and Tukey post hoc test. aShows significant difference compared to KSOMAA no stimulus control (p < 0.05). bShows significant difference compared with CC, KSOMAA, and Met + Asa + CC (p < 0.05)

The severity of effects of AMPK activity modulation is related to culture with AMPK agonists or antagonists and affects cell number. We next tested whether embryos showed immediate, morphological signs of a response to AMPK activity modulators after 1 day of culture. All treatment groups were translucent, except embryos in the CC which were slightly opaque (Fig. 4a). Embryos at the initiation of treatment at day 1 were at the two-cell stage, and cell number at day 2 suggests that the two agonists most severely limited cell proliferation and that the antagonist slightly increased proliferation compared with control or the group where antagonist was added to two agonists (Met + Asa + CC). This suggests that AMPK agonism may slow growth and AMPK antagonism speed growth. Importantly, the embryos were translucent after 1 day of treatment on day 2, and thus, DS Asa and Met did not have AMPK agonist or other moieties that immediately caused necrosis or morbidity. But, by day 4, the cells/embryo of two agonist treatment groups was not significantly different from those with the same treatment at day 2 (p > 0.05) (Fig. 4b), and these embryos were mostly dead by day 4. In contrast, the other three groups had developed higher cell number and had progressed to over 60 % blastocysts in all of these three groups. However, the CC-treated group was not highly cavitated and remained opaque. The Met + Asa + CC-treated embryos were highly cavitated but had smaller ICMs than the control group cultured in KSOMAA alone. It should be noted that the x-axis assignment of severity of effect was multifactorial and based on outcomes from days 2 and 4 and from morphological criteria and cell number. For example, CC had the highest cell number/embryo at day 2 but was also opaque, and opacity is a known predictor of low embryo quality in IVF [66]. Additionally, although progression and size of cavitation were similar at day 4 for KSOMAA and CC alone, CC alone had a smaller ICM, and these would be considered of lower quality and of lesser fitness for reimplantation in IVF [67].

Fig. 4.

Fig. 4

After 1 day, embryos in all stimuli are translucent, but development is delayed in agonists BR-DIM or Met + Asa, but by 4 days, the two agonist treatment groups have arrested with similar cell number as after 1 day (a). Embryos were stimulated on day 1 and micrographed on days 2 and 4, and cell counts were performed on day 2 embryos (white inset numbers). The severity of outcomes at day 2 (measured by cell number and opacity) and day 4 (measured by morbidity, arrest, cavitation, and ICM density) (b). Cell counts ± SEM are shown for all six stimulus groups on day 2 and for the two AMPK agonist-only stimuli (BR-DIM and Met + Asa), where nearly all embryo remained in cell countable cleavage stages, cell counts are also shown for day 4. Biological experiments were done in triplicate, and quantitative immunofluorescence of nuclei was done using Simple PCI DN module and analyzed for significance using ANOVA and Tukey post hoc test. aShows a significant difference compared to KSOMAA (p < 0.05). bShows no significant difference between day 2 and day 4 for BR-DIM and Met + Asa, but significant difference compared with KSOMAA (p < 0.05). cShows no significance compared with KSOMAA

The block of development by Met + Asa or BR-DIM suggested that Asa and BR-DIM should also be tested for AMPK-dependent potency loss at the two-cell stage. Using the same culture and assay strategy as in Fig. 1, we found that both BR-DIM and Asa cause a significant decrease in Oct4 nuclear protein in two-cell embryos (p < 0.05) (Fig. 5a). Similarly to Met alone, Asa- and BR-DIM-induced loss was significantly reversed by CC (p < 0.05) (Fig. 5b). In the case of Asa, reversal was to an Oct4 level not significantly different than unstressed two-cell embryos, suggesting a strong AMPK component in Asa effects. Interestingly, CC significantly increased Oct4 (p < 0.05) (Fig. 5a, b), suggesting that culture stress causes AMPK-dependent decrease in Oct4.

Fig. 5.

Fig. 5

AMPK mediates BR-DIM- and Asa-induced loss of nuclear Oct4 potency factor proteins that is largely reversed by CC (a). Zygotes were cultured overnight in lowest-stress media, some two-cell embryos were then preloaded with 5 μM CC for 2 h, and at time 0, embryos were incubated with 20 μM BR-DIM or 10 μM Asa ± CC or continued with CC alone for 1 h. Embryos were fixed, quenched, permeabilized, and exposed to monoclonal anti-Oct4 antibodies and counterstained with anti-mouse FITC and Hoechst and then micrographed. Embryos were treated with KSOMAA alone (A, B), 5 μM CC alone (C, D), 20 μM BR-DIM alone (E, F), BR-DIM + CC (G, H), 10 μM Asa (I, J), and Asa + CC (K, L). The micrographs represent Hoechst (A, C, E, G, I, K) and Oct4 (B, D, F, H, J, L) after 1 h of the indicated treatments after exposures of the same embryos (b). AMPK induces loss of nuclear potency factors through BR-DIM or Asa, and loss is reversed by AMPK antagonist CC. Biological experiments were done in triplicate, and quantitative immunofluorescence of nuclei was done using Simple PCI DN module and analyzed for significance using ANOVA and Tukey post hoc test. aShows a significant difference compared with KSOMAA control (p < 0.05). bShows a significant difference between CC and KSOMAA, BR-DIM, and BR-DIM + CC or between Asa and Asa + CC and a significant difference compared with KSOMAA (p < 0.05). cShows significant difference between Asa + CC compared with Asa (p < 0.05) but no significant difference compared with KSOMAA

Discussion

For the first time, we show here that AMPK agonist drugs, Asa and Met, and DS BR-DIM can have negative effects on stem cell potency, cell growth, and embryo development in early mammalian development. Cultured embryos are translucent and not immediately morbid after 1 day of culture with the two AMPK agonist treatments Met + Asa or BR-DIM. But, cell growth is retarded and rapidly arrested and cell accumulation is highly decreased compared with media control. The AMPK antagonist CC improves slowed cell growth and early retardation of embryo development by AMPK agonists in the first day of treatment (e.g., day 2). But, full effects on reversal of retardation are not apparent until the third and last day of treatment in culture (e.g., day 4). It is clear that CC reverses the effects of Met + Asa or BR-DIM, but it is likely that some of the agonists or the antagonists are having their effects solely through AMPK.

We sought to test the hypothesis that many stimuli cause AMPK-dependent ESC potency factor loss in two-cell embryo, as had been shown for TSCs, blastocysts, and TSC potency factors in two-cell embryos. For the first time, we arrested development by the blastocyst stagearrested development by the blastocyst stagearrested development by the blastocyst stagearrested development by the blastocyst stage cause AMPK-dependent Oct4 and Rex1 potency factor loss in two-cell embryos as Met-, Asa-, or BR-DIM-induced loss is largely reversed by the AMPK antagonist CC. Interestingly, the rapid potency loss at 1 h occurs in two-cell embryos and Met + Asa or BR-DIM delays or stops embryonic development at the two-cell stage or soon after. Taken together, these data suggest that rapid potency factor loss could be part of the mechanism of embryo delay. But, AMPK also is known to mediate anabolic to catabolic metabolism shifts in oocytes, embryos, and stem cells from the embryos [22, 24, 41], and this hypothetically would mediate delay. An important aspect of future studies is to test for AMPK-dependent effects on decreasing anabolism in cultured embryos.

AMPK agonists have been reported to enable cultured oocytes stressed by four different stressors to mature [22], or oocytes or blastocysts derived from metabolically stressed diabetic mothers [23, 25], to develop more normally than occurs in culture with media alone. Met improves maternal metabolism [6, 6876] and ovulation [68, 77] and is good for oocytes and embryos derived from females under obese and diabetic conditions [2325, 78], and thus, AMPK can improve compromised oocytes and embryos. CC alone increases Oct4, suggesting that there is some stress during culture in optimal KSOM media and high clinical doses of single and paired AMPK agonists have much larger effect on potency loss on near-normal embryos. The results here do not contradict the reports of positive functions for AMPK in gametogenesis and embryogenesis on specimens derived from or in conditions of stress. Our data suggest that potency is lost and morbidity increases when levels of AMPK activity are above those in normal, low-stress embryos cultured in low-stress media, or when metabolically stressed embryos are treated by increasing abnormally low AMPK [22, 25] activity back to an optimum level.

We used hyperosmotic sorbitol at 200 mM because this dose is not toxic to embryos, TSCs, or ESCs [41, 55, 56, 65, 79] but slows their proliferation and has significant effects in causing AMPK-dependent Cdx2 and Id2 loss [41, 45], PL1 increase in TSCs [80], Oct4 and Rex1 loss and first lineage Dab2 and LRP2 markers in ESCs [65, 81, 82], and significant AMPK-dependent Id2 loss in blastocysts [41]. We had previously shown that 200 mM sorbitol causes Cdx2 and Id2 loss in two-cell embryos in an AMPK-dependent manner [45], and this report adds AMPK-dependent Rex1 and Oct4 loss to make up a group of four AMPK-regulated potency factors expressed in both ESC and TSC lineages by the blastocyst stage. When taken together with BR-DIM, Asa, or Met, all purative AMPK agonist stimuli tested to date mediate AMPK-dependent potency loss in early embryos. The single AMPK antagonist tested, CC, increased potency.

Although potency factors may begin to prime cells in the four-cell embryo [83] to allocate later to the two stem cell lineages at the blastocyst stage, AMPK-mediated loss of both ESC and TSC lineage potency factors does not yet indicate that AMPK is a regulating potency that favors either lineage at the two- to four-cell stage.

Do Asa and other drugs get to the uterine fluids? Early studies of radioactive drugs suggest that Asa is at the same level in uterine fluids as plasma [84, 85]. Although Asa was not studied, many drugs such as barbital, caffeine, and nicotine are taken up by the gestationally exposed blastocysts [86]. A cocktail of small compounds, including salicylate, led to gestational exposure and ensuing intrauterine growth restriction (IUGR) [87], although individually, only nicotine and DDT caused IUGR. Asa is associated with miscarriage after early pregnancy exposure [88].

The 10 μM Asa dose used here was well within normal usage where normal to medium doses produce Cmax plasma doses of ~2–200 μM [61, 89]. Interestingly, it was previously reported that Asa blocks development of cultured embryos to the blastocyst stage and reduces the number of cells/embryo [90, 91]. Although an Asa-only test for embryo development blockage was not done here, the Met + Asa and BR-DIM treatments are consistent with this and, for the first time, show an AMPK dependence of the arrest.

These Asa preimplantation studies of the 1950s and 1960s were done before Met was studied, but Met is a small molecule like Asa and is likely to enter follicular and luminal fluids. Recent studies have shown that Met is significantly higher in umbilical arterial and venous plasma than in maternal plasma [92], suggesting that Met crosses the placenta and accesses the embryo and its stem cells after implantation. The earliest post-implantation human embryo in the trophoblast plate stage and lacunar stage has direct access to maternal capillary plasma, which surrounds the embryo [93, 94].

The Met dose used here was 1000 μM, the same one used to study non-morbid regulation of the transcription factor Pax3 during a 3-day culture of ESCs [58]. Here, this dose caused Oct4 and Rex1 loss that was reversed largely by CC, suggesting AMPK dependence. Using a high dose such as 1 mM, when reversed by CC, suggests strongly that Asa effects are through AMPK and not off-target effects, which would be highest at high doses. With the addition of 10 μM Asa, metformin also blocked embryo development, soon after addition of the two drugs at the two-cell stage. Ultimately, this caused an 95 % block of development to the blastocyst stage, Interestingly, it was previously shown that a 780 μM metformin caused a 93 % block of blastocyst development from the two-cell stage [95], similar to the 95 % block by 1000 μM Met and 10 μM Asa here. In these previous studies, the normal plasma dose of 39 μM had no effect on blocking development blastocyst from two-cell embryos during culture. Our next hypothesis to be tested in future studies is that combined exposure to normal plasma levels of 39 μM Met and 10 μM Asa will block embryo development in vitro and in vivo as Met and Asa are known to synergize in activating AMPK on rates and deactivating AMPK off rates, respectively [39].

The 20-μM dose for BR-DIM used here is above the 2.5-μm therapeutic plasma dose range for treatments of cervical dysplasia [18] and dose range used for anti-cancer treatments that slow growth and can kill prostate cancer cells [96]. However, different diet formulations can increase BR-DIM to 7 μM in plasma, and the anti-cancer uses of BR-DIM have led to new formulations and dietary combinations (e.g., delivery with cod liver oil) that will improve absorption and enable a 20-μM plasma dose. Here, 20 μM BR-DIM caused highest loss of nuclear Oct4 potency factor protein compared with all other stimuli. This loss was reversed by CC, suggesting AMPK-dependent loss. The same BR-DIM dose quickly arrests all two-cell embryos after 24-h exposure, although these initially appear healthy. By the blastocyst stage, 2 days later, the BR-DIM-treated embryos had not significantly increased cell number but were opaque and dead. Co-addition of CC with BR-DIM increased the fraction of embryos reaching blastocyst stage from 60 % compared with 0 % blastocysts for BR-DIM alone.

These data clearly show that BR-DIM is the most powerful blocker of Oct4, causing nearly 85 % loss, and of development of culture cell embryos to blastocyst stage, causing 100 % block. CC significantly but not totally reverses both effects. This suggests that BR-DIM can act powerfully on the early embryo and may do so through AMPK-dependent and AMPK-independent mechanisms.

It is likely that Met, Asa, and BR-DIM reach plasma and uterine fluid levels that would stimulate AMPK-dependent responses in vivo as well as in vitro. However, Met [97], Asa [98], and BR-DIM [60, 99] reach pharmacokinetic peaks within 30–144 min of ingestion but are cleared within 12 h. It will be important to expose gestational females to these drugs and remove embryos at the timing of the peak to test for potency factor loss. We previously showed that shear stress by mouth pipetting causes activation of stress-activated protein kinase (SAPK) to levels as high as highest activation dose of hyperosmotic sorbitol, about a 10-fold increase [30]. However, this dose has no effect of mouth pipetting for 15 min on cell number 24 h later [100]. Thus, transient high molecular responses may not lead to biological changes. In addition, three of four ESC potency factors that were decreased from 1 to 4 h of 200 mM sorbitol rebounded to normal levels at 24 h despite continuing hyperosmotic stress [65]. These data suggest that in vitro effects may be higher due to lack of clearance and that ESCs in the early embryo may regain potency after prolonged exposures. Either hypothesis requires testing in vivo.

If results here are corroborated in vivo, then some caution should be used when prescribing Met for PCOS and diabetic women, especially after potential sensitive periods of ovulation, fertilization, and rapid growth and differentiation starting at the blastocyst stage that produces anti-luteolytic hormones. Some embryo loss was noted in women treated with Met alone after detection of chemical pregnancy and before clinical detection by ultrasound [12]. But, in general, the use of Met improves pregnancy compared with untreated women with lessened infertility where the oocytes and blastocysts are compromised and many causes of infertility lessen AMPK activity [11, 101, 102]. However, the possibility of additive or synergistic exposure of drugs and DSs with AMPK agonist activity deserves further study in vitro and in vivo. This is especially true since AMPK agonists can both activate the enzyme through the positive regulatory Ύ-subunit and by inactivating the negative regulatory β-subunit, causing supramaximal effects [38, 39, 103].

Similar to past reports for two-cell embryos [45], blastocysts [41, 45], TSCs [41, 45, 46, 63, 104, 105], ESCs [41, 64, 65], and induced pluripotent stem cells (iPSCs) [43], AMPK is the master regulatory enzyme for stress-induced potency loss of Oct4, Rex1, Cdx1, Id2, [41, 4346, 106], and Nanog [42]. Oct4 and Rex1 and other potency factor loss in two-cell-stage embryos, blastocysts, TSCs, and ESCs occur within 0.5–2 h, suggesting that the stemness program change may be an important strategy requiring rapid, molecular action.

However, it is not clear what the stress-induced, AMPK-dependent Id2, Cdx2, Oct4, or Rex1 loss effects are in early embryos. For TSCs, it is clear that dominant negative Id2 loss is needed to enable stem cell differentiation to produce anti-luteolytic placental lactogen (PL)1 in rodents [41, 80]. In the two-cell-stage embryo, the role of Oct4 and other potency factors is not established. Oct4 may be needed for zygotic genome activation in zebrafish [107, 108], and the authors suggest that conservation of binding sites suggests similar roles in mammals. Oct4 and Cdx2 are necessary at the blastocyst stage to allocate ESC and TSC lineages, respectively [109], and knockouts for either of these are lethal at the blastocyst stage [51, 110]. But, Oct4 also regulates stem cell metabolism [50] and is necessary for stress preparation and response [48, 49]. But, in mouse and human ESCs, Oct4 actively silences hundreds of promoters [111, 112] that would mediate differentiation. After knockdown of maternal and zygotic Cdx2, the most highly upregulated mRNA in mouse blastocysts is Hand1 [113], a marker of first differentiated lineage of TSC [32]. This suggests that like Oct4, Cdx2 function may also contribute to silencing possible differentiated lineages. There are many candidate mechanisms that may be dysregulated soon after addition at the two-cell-stage embryo, when AMPK agonists mediate loss of these potency factors. Although death is delayed, the early arrest soon after the two-cell stage suggests that AMPK-regulated molecular mechanisms at this stage are the best candidates for pathogenicity.

Many food groups and DSs are agonists for AMPK. The following food groups and food extracts have high AMPK-activating capacity: resveratrol (red grape skin extract) [114118], nootkatone (grapefruit extract) [119], ajoene (garlic extract) [120], BDIM (cruciferous vegetable extract) [19, 121], and many others [122]. Medicines activate AMPK including Asa [16], Met [35], phenformin [123, 124], rosiglitazone, and phenobarbital [125, 126], and there may be others.

Taken together, the data reported here suggest the need for further studies testing for effects in vivo for stimuli reported here. Further studies using in vitro high-throughput screening methods for stress-induced potency loss [64] are needed to test for single and additive AMPK agonists from diet and drug sources that cause potency loss and diminish stem cell growth. Since no single hypothetical mechanism for AMPK-mediated pathogenicity stands out alone, studies of changes in the global transcriptome, epigenome, and metabolome of cultured embryos are needed to help sharpen hypotheses of mechanisms.

Acknowledgments

Thanks to Jose Cibelli, Michael Diamond, and Erica Louden for comments on the manuscript.

Compliance with ethical standards

Funding

DAR and EEP from the Office of the Vice President for Research at Wayne State University, from the REI fellows’ fund (AB), and from the funding of the Mary Iacobell and Kamran Moghissi Endowed Chairs.

Footnotes

Capsule

Drugs metformin and aspirin and diet supplement BR-DIM cause AMPK-dependent potency loss and decrease embryonic development from two-cell to blastocyst stage.

References

  • 1.Duranteau L, Lefevre P, Jeandidier N, Simon T, Christin-Maitre S. Should physicians prescribe metformin to women with polycystic ovary syndrome PCOS? Ann Endocrinol (Paris) 2010;71:25–7. doi: 10.1016/j.ando.2009.12.005. [DOI] [PubMed] [Google Scholar]
  • 2.Palomba S, Pasquali R, Orio F, Jr, Nestler JE. Clomiphene citrate, metformin or both as first-step approach in treating anovulatory infertility in patients with polycystic ovary syndrome (PCOS): a systematic review of head-to-head randomized controlled studies and meta-analysis. Clin Endocrinol (Oxf) 2009;70:311–21. doi: 10.1111/j.1365-2265.2008.03369.x. [DOI] [PubMed] [Google Scholar]
  • 3.Sinawat S, Buppasiri P, Lumbiganon P, Pattanittum P. Long versus short course treatment with metformin and clomiphene citrate for ovulation induction in women with PCOS. Cochrane Database Syst Rev 2008:CD006226. [DOI] [PubMed]
  • 4.Jamal A, Milani F, Al-Yasin A. Evaluation of the effect of metformin and aspirin on utero placental circulation of pregnant women with PCOS. Iran J Reprod Med. 2012;10:265–70. [PMC free article] [PubMed] [Google Scholar]
  • 5.de Oliveira Baraldi C, Lanchote VL, de Jesus Antunes N, de Jesus Ponte Carvalho TM, Dantas Moises EC, Duarte G, et al. Metformin pharmacokinetics in nondiabetic pregnant women with polycystic ovary syndrome. Eur J Clin Pharmacol. 2011;67:1027–33. doi: 10.1007/s00228-011-1053-0. [DOI] [PubMed] [Google Scholar]
  • 6.Vause TD, Cheung AP, Sierra S, Claman P, Graham J, Guillemin JA, et al. Ovulation induction in polycystic ovary syndrome. J Obstet Gynaecol Can. 2010;32:495–502. doi: 10.1016/S1701-2163(16)34504-2. [DOI] [PubMed] [Google Scholar]
  • 7.Jungheim ES, Odibo AO. Fertility treatment in women with polycystic ovary syndrome: a decision analysis of different oral ovulation induction agents. Fertil Steril. 2010;94:2659–64. doi: 10.1016/j.fertnstert.2010.03.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Genazzani AD, Ricchieri F, Lanzoni C. Use of metformin in the treatment of polycystic ovary syndrome. Womens Health (Lond Engl) 2010;6:577–93. doi: 10.2217/whe.10.43. [DOI] [PubMed] [Google Scholar]
  • 9.Palomba S, Falbo A, Russo T, Orio F, Tollino A, Zullo F. Role of metformin in patients with polycystic ovary syndrome: the state of the art. Minerva Ginecol. 2008;60:77–82. [PubMed] [Google Scholar]
  • 10.Escobar-Morreale HF. Polycystic ovary syndrome: treatment strategies and management. Expert Opin Pharmacother. 2008;9:2995–3008. doi: 10.1517/14656560802559932. [DOI] [PubMed] [Google Scholar]
  • 11.Moll E, van der Veen F, van Wely M. The role of metformin in polycystic ovary syndrome: a systematic review. Hum Reprod Update. 2007;13:527–37. doi: 10.1093/humupd/dmm026. [DOI] [PubMed] [Google Scholar]
  • 12.Legro RS, Barnhart HX, Schlaff WD, Carr BR, Diamond MP, Carson SA, et al. Clomiphene, metformin, or both for infertility in the polycystic ovary syndrome. N Engl J Med. 2007;356:551–66. doi: 10.1056/NEJMoa063971. [DOI] [PubMed] [Google Scholar]
  • 13.Cheang KI, Sharma ST, Nestler JE. Is metformin a primary ovulatory agent in patients with polycystic ovary syndrome? Gynecol Endocrinol. 2006;22:595–604. doi: 10.1080/09513590601005847. [DOI] [PubMed] [Google Scholar]
  • 14.Carrington B, Sacks G, Regan L. Recurrent miscarriage: pathophysiology and outcome. Curr Opin Obstet Gynecol. 2005;17:591–7. doi: 10.1097/01.gco.0000194112.86051.26. [DOI] [PubMed] [Google Scholar]
  • 15.Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. 2001;108:1167–74. doi: 10.1172/JCI13505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hawley SA, Fullerton MD, Ross FA, Schertzer JD, Chevtzoff C, Walker KJ, et al. The ancient drug salicylate directly activates AMP-activated protein kinase. Science. 2012;336:918–22. doi: 10.1126/science.1215327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Higdon JV, Delage B, Williams DE, Dashwood RH. Cruciferous vegetables and human cancer risk: epidemiologic evidence and mechanistic basis. Pharmacol Res. 2007;55:224–36. doi: 10.1016/j.phrs.2007.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Del Priore G, Gudipudi DK, Montemarano N, Restivo AM, Malanowska-Stega J, Arslan AA. Oral diindolylmethane (DIM): pilot evaluation of a nonsurgical treatment for cervical dysplasia. Gynecol Oncol. 2010;116:464–7. doi: 10.1016/j.ygyno.2009.10.060. [DOI] [PubMed] [Google Scholar]
  • 19.Ribaux P, Irion O, Cohen M. An active product of cruciferous vegetables, 3,3'-diindolylmethane, inhibits invasive properties of extravillous cytotrophoblastic cells. Neuro Endocrinol Lett. 2012;33:133–7. [PubMed] [Google Scholar]
  • 20.Cohen JH, Kristal AR, Stanford JL. Fruit and vegetable intakes and prostate cancer risk. J Natl Cancer Inst. 2000;92:61–8. doi: 10.1093/jnci/92.1.61. [DOI] [PubMed] [Google Scholar]
  • 21.Chen D, Banerjee S, Cui QC, Kong D, Sarkar FH, Dou QP. Activation of AMP-activated protein kinase by 3,3'-diindolylmethane (DIM) is associated with human prostate cancer cell death in vitro and in vivo. PLoS ONE. 2012;7 doi: 10.1371/journal.pone.0047186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.LaRosa C, Downs SM. Stress stimulates AMP-activated protein kinase and meiotic resumption in mouse oocytes. Biol Reprod. 2006;74:585–92. doi: 10.1095/biolreprod.105.046524. [DOI] [PubMed] [Google Scholar]
  • 23.Louden ED, Luzzo KM, Jimenez PT, Chi T, Chi M, Moley KH. TallyHO obese female mice experience poor reproductive outcomes and abnormal blastocyst metabolism that is reversed by metformin. Reprod Fertil Dev. 2014;27:31–9. doi: 10.1071/RD14339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Louden E, Chi MM, Moley KH. Crosstalk between the AMP-activated kinase and insulin signaling pathways rescues murine blastocyst cells from insulin resistance. Reproduction. 2008;136:335–44. doi: 10.1530/REP-08-0161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ratchford AM, Chang AS, Chi MM, Sheridan R, Moley KH. Maternal diabetes adversely affects AMP-activated protein kinase activity and cellular metabolism in murine oocytes. Am J Physiol Endocrinol Metab. 2007;293:E1198–206. doi: 10.1152/ajpendo.00097.2007. [DOI] [PubMed] [Google Scholar]
  • 26.Solano ME, Elia E, Luchetti CG, Sander V, Di Girolamo G, Gonzalez C, et al. Metformin prevents embryonic resorption induced by hyperandrogenisation with dehydroepiandrosterone in mice. Reprod Fertil Dev. 2006;18:533–44. doi: 10.1071/RD05099. [DOI] [PubMed] [Google Scholar]
  • 27.Larosa C, Downs SM. Meiotic induction by heat stress in mouse oocytes: involvement of AMP-activated protein kinase and MAPK family members. Biol Reprod. 2007;76:476–86. doi: 10.1095/biolreprod.106.057422. [DOI] [PubMed] [Google Scholar]
  • 28.Chen J, Hudson E, Chi MM, Chang AS, Moley KH, Hardie DG et al. AMPK regulation of mouse oocyte meiotic resumption in vitro. Dev Biol 2006. [DOI] [PubMed]
  • 29.Rappolee DA. Impact of transient stress and stress enzymes on development. Dev Biol. 2007;304:1–8. doi: 10.1016/j.ydbio.2006.12.032. [DOI] [PubMed] [Google Scholar]
  • 30.Puscheck EE, Awonuga AO, Yang Y, Jiang Z, Rappolee DA. Molecular biology of the stress response in the early embryo and its stem cells. Adv Exp Med Biol. 2015;843:77–128. doi: 10.1007/978-1-4939-2480-6_4. [DOI] [PubMed] [Google Scholar]
  • 31.Mansouri L, Xie Y, Rappolee DA. Adaptive and pathogenic responses to stress by stem cells during development. Cells. 2012;1:1197–224. doi: 10.3390/cells1041197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Xie Y, Awonuga AO, Zhou S, Puscheck EE, Rappolee DA. Interpreting the stress response of early mammalian embryos and their stem cells. Int Rev Cell Mol Biol. 2011;287:43–95. doi: 10.1016/B978-0-12-386043-9.00002-5. [DOI] [PubMed] [Google Scholar]
  • 33.Hardie DG. Minireview: the AMP-activated protein kinase cascade: the key sensor of cellular energy status. Endocrinology. 2003;144:5179–83. doi: 10.1210/en.2003-0982. [DOI] [PubMed] [Google Scholar]
  • 34.Winder WW, Hardie DG. AMP-activated protein kinase, a metabolic master switch: possible roles in type 2 diabetes. Am J Physiol. 1999;277:E1–10. doi: 10.1152/ajpendo.1999.277.1.E1. [DOI] [PubMed] [Google Scholar]
  • 35.Hardie DG. Neither LKB1 nor AMPK are the direct targets of metformin. Gastroenterology. 2006;131:973. doi: 10.1053/j.gastro.2006.07.032. [DOI] [PubMed] [Google Scholar]
  • 36.Fryer LG, Parbu-Patel A, Carling D. The Anti-diabetic drugs rosiglitazone and metformin stimulate AMP-activated protein kinase through distinct signaling pathways. J Biol Chem. 2002;277:25226–32. doi: 10.1074/jbc.M202489200. [DOI] [PubMed] [Google Scholar]
  • 37.Yue W, Wang T, Zachariah E, Lin Y, Yang CS, Xu Q, et al. Transcriptomic analysis of pancreatic cancer cells in response to metformin and aspirin: an implication of synergy. Sci Rep. 2015;5:13390. doi: 10.1038/srep13390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yue W, Yang CS, DiPaola RS, Tan XL. Repurposing of metformin and aspirin by targeting AMPK-mTOR and inflammation for pancreatic cancer prevention and treatment. Cancer Prev Res. 2014;7:388–97. doi: 10.1158/1940-6207.CAPR-13-0337. [DOI] [PubMed] [Google Scholar]
  • 39.Ford RJ, Fullerton MD, Pinkosky SL, Day EA, Scott JW, Oakhill JS, et al. Metformin and salicylate synergistically activate liver AMPK, inhibit lipogenesis and improve insulin sensitivity. Biochem J. 2015;468:125–32. doi: 10.1042/BJ20150125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Downs SM, Hudson ER, Hardie DG. A potential role for AMP-activated protein kinase in meiotic induction in mouse oocytes. Dev Biol. 2002;245:200–12. doi: 10.1006/dbio.2002.0613. [DOI] [PubMed] [Google Scholar]
  • 41.Zhong W, Xie Y, Abdallah M, Awonuga AO, Slater JA, Sipahi L, et al. Cellular stress causes reversible, PRKAA1/2-, and proteasome-dependent ID2 protein loss in trophoblast stem cells. Reproduction. 2010;140:921–30. doi: 10.1530/REP-10-0268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chae HD, Lee MR, Broxmeyer HE. 5-Aminoimidazole-4-carboxyamide ribonucleoside induces G(1)/S arrest and Nanog downregulation via p53 and enhances erythroid differentiation. Stem Cells. 2012;30:140–9. doi: 10.1002/stem.778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Vazquez-Martin A, Vellon L, Quiros PM, Cufi S, Ruiz de Galarreta E, Oliveras-Ferraros C, et al. Activation of AMP-activated protein kinase (AMPK) provides a metabolic barrier to reprogramming somatic cells into stem cells. Cell Cycle. 2012;11:974–89. doi: 10.4161/cc.11.5.19450. [DOI] [PubMed] [Google Scholar]
  • 44.Yang Y, Jiang Z, Bolnick A, Dai J, Puscheck E, Rappolee D. Departure from optimal 2% O2 level for TSC potency and proliferation leads to most rapid increases in AMPK activity. Submitted to Placenta 2015. [DOI] [PMC free article] [PubMed]
  • 45.Xie Y, Awonuga A, Liu J, Rings E, Puscheck EE, Rappolee DA. Stress induces AMPK-dependent loss of potency factors Id2 and Cdx2 in early embryos and stem cells [corrected] Stem Cells Dev. 2013;22:1564–75. doi: 10.1089/scd.2012.0352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Xie Y, Abdallah ME, Awonuga AO, Slater JA, Puscheck EE, Rappolee DA. Benzo(a)pyrene causes PRKAA1/2-dependent ID2 loss in trophoblast stem cells. Mol Reprod Dev. 2010;77:533–9. doi: 10.1002/mrd.21178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Guo Y, Mantel C, Hromas RA, Broxmeyer HE. Oct-4 is critical for survival/antiapoptosis of murine embryonic stem cells subjected to stress: effects associated with Stat3/survivin. Stem Cells. 2008;26:30–4. doi: 10.1634/stemcells.2007-0401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kang J, Shakya A, Tantin D. Stem cells, stress, metabolism and cancer: a drama in two Octs. Trends Biochem Sci. 2009;34:491–9. doi: 10.1016/j.tibs.2009.06.003. [DOI] [PubMed] [Google Scholar]
  • 49.Kang J, Gemberling M, Nakamura M, Whitby FG, Handa H, Fairbrother WG, et al. A general mechanism for transcription regulation by Oct1 and Oct4 in response to genotoxic and oxidative stress. Genes Dev. 2009;23:208–22. doi: 10.1101/gad.1750709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Frum T, Halbisen MA, Wang C, Amiri H, Robson P, Ralston A. Oct4 cell-autonomously promotes primitive endoderm development in the mouse blastocyst. Dev Cell. 2013;25:610–22. doi: 10.1016/j.devcel.2013.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nichols J, Zevnik B, Anastassiadis K, Niwa H, Klewe-Nebenius D, Chambers I, et al. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell. 1998;95:379–91. doi: 10.1016/S0092-8674(00)81769-9. [DOI] [PubMed] [Google Scholar]
  • 52.Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet. 2000;24:372–6. doi: 10.1038/74199. [DOI] [PubMed] [Google Scholar]
  • 53.Hogan B, Beddington R, Constantini F, Lacy B. Manipulating the mouse embryo: a laboratory manual. 3. Cold Spring Harbor: Cold Spring Harbor Laboratory; 2002. [Google Scholar]
  • 54.Wang Y, Puscheck EE, Lewis JJ, Trostinskaia AB, Wang F, Rappolee DA. Increases in phosphorylation of SAPK/JNK and p38MAPK correlate negatively with mouse embryo development after culture in different media. Fertil Steril. 2005;83(Suppl 1):1144–54. doi: 10.1016/j.fertnstert.2004.08.038. [DOI] [PubMed] [Google Scholar]
  • 55.Zhong W, Xie Y, Wang Y, Lewis J, Trostinskaia A, Wang F, et al. Use of hyperosmolar stress to measure stress-activated protein kinase activation and function in human HTR cells and mouse trophoblast stem cells. Reprod Sci. 2007;14:534–47. doi: 10.1177/1933719107307182. [DOI] [PubMed] [Google Scholar]
  • 56.Xie Y, Zhong W, Wang Y, Trostinskaia A, Wang F, Puscheck EE, et al. Using hyperosmolar stress to measure biologic and stress-activated protein kinase responses in preimplantation embryos. Mol Hum Reprod. 2007;13:473–81. doi: 10.1093/molehr/gam027. [DOI] [PubMed] [Google Scholar]
  • 57.An Y, Sun Z, Li L, Zhang Y, Ji H. Relationship between psychological stress and reproductive outcome in women undergoing in vitro fertilization treatment: psychological and neurohormonal assessment. J Assist Reprod Genet. 2013;30:35–41. doi: 10.1007/s10815-012-9904-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lee HY, Wei D, Loeken MR. Lack of metformin effect on mouse embryo AMPK activity: implications for metformin treatment during pregnancy. Diabetes Metab Res Rev. 2014;30:23–30. doi: 10.1002/dmrr.2451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wu Y, Viana M, Thirumangalathu S, Loeken MR. AMP-activated protein kinase mediates effects of oxidative stress on embryo gene expression in a mouse model of diabetic embryopathy. Diabetologia. 2012;55:245–54. doi: 10.1007/s00125-011-2326-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Paltsev M, Kiselev V, Muyzhnek E, Drukh V, Kuznetsov I, Pchelintseva O. Comparative preclinical pharmacokinetics study of 3,3'-diindolylmethane formulations: is personalized treatment and targeted chemoprevention in the horizon? EPMA J. 2013;4:25. doi: 10.1186/1878-5085-4-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ross-Lee LM, Elms MJ, Cham BE, Bochner F, Bunce IH, Eadie MJ. Plasma levels of aspirin following effervescent and enteric coated tablets, and their effect on platelet function. Eur J Clin Pharmacol. 1982;23:545–51. doi: 10.1007/BF00637504. [DOI] [PubMed] [Google Scholar]
  • 62.Hundal RS, Petersen KF, Mayerson AB, Randhawa PS, Inzucchi S, Shoelson SE, et al. Mechanism by which high-dose aspirin improves glucose metabolism in type 2 diabetes. J Clin Invest. 2002;109:1321–6. doi: 10.1172/JCI0214955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Yang Y, Arenas-Hernandez M, Gomez-Lopez N, Dai J, Puscheck E, Rappolee D. Hypoxic stress forces large, irreversible trophoblast stem cell differentiation. Biol Reprod. 2016;Submitted. [DOI] [PMC free article] [PubMed]
  • 64.Li Q, Gomez-Lopez N, Drewlo S, Sanchez-Rodriquez E, Dai J, Puscheck EE et al. Development and validation of a Rex1-RFP potency activity reporter assay that quantifies stress-forced potency loss in mouse embryonic stem cells. Stem Cells Dev 2015. [DOI] [PMC free article] [PubMed]
  • 65.Slater JA, Zhou S, Puscheck EE, Rappolee DA. Stress-induced enzyme activation primes murine embryonic stem cells to differentiate toward the first extraembryonic lineage. Stem Cells Dev. 2014;23:3049–64. doi: 10.1089/scd.2014.0157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.McRae AC, Church RB. Cytoplasmic projections of trophectoderm distinguish implanting from preimplanting and implantation-delayed mouse blastocytes. J Reprod Fertil. 1990;88:31–40. doi: 10.1530/jrf.0.0880031. [DOI] [PubMed] [Google Scholar]
  • 67.Shapiro BS, Daneshmand ST, Garner FC, Aguirre M, Hudson C. Freeze-all at the blastocyst or bipronuclear stage: a randomized clinical trial. Fertil Steril. 2015;104:1138–44. doi: 10.1016/j.fertnstert.2015.07.1141. [DOI] [PubMed] [Google Scholar]
  • 68.Rice S, Elia A, Jawad Z, Pellatt L, Mason HD. Metformin inhibits follicle-stimulating hormone (FSH) action in human granulosa cells: relevance to polycystic ovary syndrome. J Clin Endocrinol Metab. 2013;98:E1491–500. doi: 10.1210/jc.2013-1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kovacs G. How to improve your ART success rates: an evidence-based review of adjuncts to IVF. Cambridge. New York: Cambridge University Press; 2011. [Google Scholar]
  • 70.Palomba S, Orio F, Jr, Russo T, Falbo A, Cascella T, Colao A, et al. Is ovulation induction still a therapeutic problem in patients with polycystic ovary syndrome? J Endocrinol Invest. 2004;27:796–805. doi: 10.1007/BF03347527. [DOI] [PubMed] [Google Scholar]
  • 71.Kashyap S, Wells GA, Rosenwaks Z. Insulin-sensitizing agents as primary therapy for patients with polycystic ovarian syndrome. Hum Reprod. 2004;19:2474–83. doi: 10.1093/humrep/deh440. [DOI] [PubMed] [Google Scholar]
  • 72.Ben-Haroush A, Yogev Y, Fisch B. Insulin resistance and metformin in polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol. 2004;115:125–33. doi: 10.1016/j.ejogrb.2003.11.027. [DOI] [PubMed] [Google Scholar]
  • 73.Barbieri RL. Metformin for the treatment of polycystic ovary syndrome. Obstet Gynecol. 2003;101:785–93. doi: 10.1016/s0029-7844(03)00045-0. [DOI] [PubMed] [Google Scholar]
  • 74.Nestler JE, Stovall D, Akhter N, Iuorno MJ, Jakubowicz DJ. Strategies for the use of insulin-sensitizing drugs to treat infertility in women with polycystic ovary syndrome. Fertil Steril. 2002;77:209–15. doi: 10.1016/S0015-0282(01)02963-6. [DOI] [PubMed] [Google Scholar]
  • 75.Phipps WR. Polycystic ovary syndrome and ovulation induction. Obstet Gynecol Clin N Am. 2001;28:165–82. doi: 10.1016/S0889-8545(05)70192-3. [DOI] [PubMed] [Google Scholar]
  • 76.Taylor R, Marsden PJ. Insulin sensitivity and fertility. Hum Fertil (Camb) 2000;3:65–9. doi: 10.1080/1464727002000198701. [DOI] [PubMed] [Google Scholar]
  • 77.Goldenberg N, Glueck CJ. Is pharmacogenomics our future? Metformin, ovulation and polymorphism of the STK11 gene in polycystic ovary syndrome. Pharmacogenomics. 2008;9:1163–5. doi: 10.2217/14622416.9.8.1163. [DOI] [PubMed] [Google Scholar]
  • 78.Chen J, Hudson E, Chi MM, Chang AS, Moley KH, Hardie DG, et al. AMPK regulation of mouse oocyte meiotic resumption in vitro. Dev Biol. 2006;291:227–38. doi: 10.1016/j.ydbio.2005.11.039. [DOI] [PubMed] [Google Scholar]
  • 79.Patel Y, Kim H, Rappolee DA. A role for hepatocyte growth factor during early postimplantation growth of the placental lineage in mice. Biol Reprod. 2000;62:904–12. doi: 10.1095/biolreprod62.4.904. [DOI] [PubMed] [Google Scholar]
  • 80.Awonuga AO, Zhong W, Abdallah ME, Slater JA, Zhou SC, Xie YF, et al. Eomesodermin, HAND1, and CSH1 proteins are induced by cellular stress in a stress-activated protein kinase-dependent manner. Mol Reprod Dev. 2011;78:519–28. doi: 10.1002/mrd.21342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Li Q, Louden E, Dai J, Furcron A, Gomez-Lopez N, Drewlo S et al. Stress forces first lineage differentiation of mouse ESCs, validation of a high throughput screen for toxicant stress. Development 2016;manuscript in preparation. [DOI] [PMC free article] [PubMed]
  • 82.Li Q, Gomez-Lopez N, Drewlo S, Sanchez-Rodriguez E, Dai J, Puscheck EE, et al. Development and validation of a Rex1-RFP potency activity reporter assay that quantifies stress-forced potency loss in mouse embryonic stem cells. Stem Cells Dev. 2016;25:320–8. doi: 10.1089/scd.2015.0169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Goolam M, Scialdone A, Graham SJ, Macaulay IC, Jedrusik A, Hupalowska A, et al. Heterogeneity in Oct4 and Sox2 targets biases cell fate in 4-cell mouse embryos. Cell. 2016;165:61–74. doi: 10.1016/j.cell.2016.01.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lutwak-Mann C, Laser H. Bicarbonate content of the blastocyst fluid and carbonic anhydrase in the pregnant rabbit uterus. Nature. 1954;173:268–9. doi: 10.1038/173268a0. [DOI] [PubMed] [Google Scholar]
  • 85.Lutwak-Mann C, Hay MF. Effect on the early embryo of agents administered to the mother. Br Med J. 1962;2:944–6. doi: 10.1136/bmj.2.5310.944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Fabro S. Penetration of chemicals into the oocyte, uterine fluid, and preimplantation blastocyst. Environ Health Perspect. 1978;24:25–9. doi: 10.1289/ehp.782425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Fabro S, McLachlan JA, Dames NM. Chemical exposure of embryos during the preimplantation stages of pregnancy: mortality rate and intrauterine development. Am J Obstet Gynecol. 1984;148:929–38. doi: 10.1016/0002-9378(84)90535-0. [DOI] [PubMed] [Google Scholar]
  • 88.Nielsen GL, Sorensen HT, Larsen H, Pedersen L. Risk of adverse birth outcome and miscarriage in pregnant users of non-steroidal anti-inflammatory drugs: population based observational study and case-control study. BMJ. 2001;322:266–70. doi: 10.1136/bmj.322.7281.266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Cerletti C, Bonati M, del Maschio A, Galletti F, Dejana E, Tognoni G, et al. Plasma levels of salicylate and aspirin in healthy volunteers: relevance to drug interaction on platelet function. J Lab Clin Med. 1984;103:869–77. [PubMed] [Google Scholar]
  • 90.Ying Y, Cai YX, Lou YJ. Effects of blastocyst deficiencies induced by aspirin treatment during preimplantation period in rats on development of embryos after implantation. Yao Xue Xue Bao. 1996;31:416–9. [PubMed] [Google Scholar]
  • 91.Ying Y, Lou YJ. Effects of preimplantation treatment with aspirin and acetaminophen on blastocyst and fetus in rats. Zhongguo Yao Li Xue Bao. 1993;14:369–72. [PubMed] [Google Scholar]
  • 92.Vanky E, Zahlsen K, Spigset O, Carlsen SM. Placental passage of metformin in women with polycystic ovary syndrome. Fertil Steril. 2005;83:1575–8. doi: 10.1016/j.fertnstert.2004.11.051. [DOI] [PubMed] [Google Scholar]
  • 93.Enders AC. Trophoblast-uterine interactions in the first days of implantation: models for the study of implantation events in the human. Semin Reprod Med. 2000;18:255–63. doi: 10.1055/s-2000-12563. [DOI] [PubMed] [Google Scholar]
  • 94.Enders AC, Lantz KC, Peterson PE, Hendrickx AG. From blastocyst to placenta: the morphology of implantation in the baboon. Hum Reprod Update. 1997;3:561–73. doi: 10.1093/humupd/3.6.561. [DOI] [PubMed] [Google Scholar]
  • 95.Bedaiwy MA, Miller KF, Goldberg JM, Nelson D, Falcone T. Effect of metformin on mouse embryo development. Fertil Steril. 2001;76:1078–9. doi: 10.1016/S0015-0282(01)02825-4. [DOI] [PubMed] [Google Scholar]
  • 96.Li Y, Wang Z, Kong D, Murthy S, Dou QP, Sheng S, et al. Regulation of FOXO3a/beta-catenin/GSK-3beta signaling by 3,3'-diindolylmethane contributes to inhibition of cell proliferation and induction of apoptosis in prostate cancer cells. J Biol Chem. 2007;282:21542–50. doi: 10.1074/jbc.M701978200. [DOI] [PubMed] [Google Scholar]
  • 97.Caille G, Lacasse Y, Raymond M, Landriault H, Perrotta M, Picirilli G, et al. Bioavailability of metformin in tablet form using a new high pressure liquid chromatography assay method. Biopharm Drug Dispos. 1993;14:257–63. doi: 10.1002/bdd.2510140308. [DOI] [PubMed] [Google Scholar]
  • 98.Levy G. Comparative pharmacokinetics of aspirin and acetaminophen. Arch Intern Med. 1981;141:279–81. doi: 10.1001/archinte.1981.00340030011003. [DOI] [PubMed] [Google Scholar]
  • 99.Anderton MJ, Manson MM, Verschoyle RD, Gescher A, Lamb JH, Farmer PB, et al. Pharmacokinetics and tissue disposition of indole-3-carbinol and its acid condensation products after oral administration to mice. Clin Cancer Res. 2004;10:5233–41. doi: 10.1158/1078-0432.CCR-04-0163. [DOI] [PubMed] [Google Scholar]
  • 100.Xie Y, Wang F, Puscheck EE, Rappolee DA. Pipetting causes shear stress and elevation of phosphorylated stress-activated protein kinase/jun kinase in preimplantation embryos. Mol Reprod Dev. 2007;74:1287–94. doi: 10.1002/mrd.20563. [DOI] [PubMed] [Google Scholar]
  • 101.Zhang H, Legro RS, Zhang J, Zhang L, Chen X, Huang H, et al. Decision trees for identifying predictors of treatment effectiveness in clinical trials and its application to ovulation in a study of women with polycystic ovary syndrome. Hum Reprod. 2010;25:2612–21. doi: 10.1093/humrep/deq210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Palomba S, Falbo A, Russo T, Orio F, Tolino A, Zullo F. Systemic and local effects of metformin administration in patients with polycystic ovary syndrome (PCOS): relationship to the ovulatory response. Hum Reprod. 2010;25:1005–13. doi: 10.1093/humrep/dep466. [DOI] [PubMed] [Google Scholar]
  • 103.O'Brien AJ, Villani LA, Broadfield LA, Houde VP, Galic S, Blandino G, et al. Salicylate activates AMPK and synergizes with metformin to reduce the survival of prostate and lung cancer cells ex vivo through inhibition of de novo lipogenesis. Biochem J. 2015;469:177–87. doi: 10.1042/BJ20150122. [DOI] [PubMed] [Google Scholar]
  • 104.Xie Y, Zhou S, Jiang Z, Dai J, Puscheck EE, Lee I, et al. Hypoxic stress induces, but cannot sustain trophoblast stem cell differentiation to labyrinthine placenta due to mitochondrial insufficiency. Stem Cell Res. 2014;13:478–91. doi: 10.1016/j.scr.2014.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhou S, Xie Y, Puscheck EE, Rappolee DA. Oxygen levels that optimize TSC culture are identified by maximizing growth rates and minimizing stress. Placenta. 2011;32:475–81. doi: 10.1016/j.placenta.2011.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Abdulhasan M, Yang Y, Dai J, Folger J, Cibelli J, Shubber A et al. CoQ10 improves bovine oocyte IVM, increases ATP and potency factor levels while maintaining decreased AMPK activity and stress marker levels. Biol Reprod. 2015;To be submitted.
  • 107.Lee MT, Bonneau AR, Takacs CM, Bazzini AA, DiVito KR, Fleming ES, et al. Nanog, Pou5f1 and SoxB1 activate zygotic gene expression during the maternal-to-zygotic transition. Nature. 2013;503:360–4. doi: 10.1038/nature12632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Leichsenring M, Maes J, Mossner R, Driever W, Onichtchouk D. Pou5f1 transcription factor controls zygotic gene activation in vertebrates. Science. 2013;341:1005–9. doi: 10.1126/science.1242527. [DOI] [PubMed] [Google Scholar]
  • 109.Niwa H, Toyooka Y, Shimosato D, Strumpf D, Takahashi K, Yagi R, et al. Interaction between Oct3/4 and Cdx2 determines trophectoderm differentiation. Cell. 2005;123:917–29. doi: 10.1016/j.cell.2005.08.040. [DOI] [PubMed] [Google Scholar]
  • 110.Strumpf D, Mao CA, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, et al. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132:2093–102. doi: 10.1242/dev.01801. [DOI] [PubMed] [Google Scholar]
  • 111.Lee TI, Jenner RG, Boyer LA, Guenther MG, Levine SS, Kumar RM, et al. Control of developmental regulators by Polycomb in human embryonic stem cells. Cell. 2006;125:301–13. doi: 10.1016/j.cell.2006.02.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Boyer LA, Plath K, Zeitlinger J, Brambrink T, Medeiros LA, Lee TI, et al. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature. 2006;441:349–53. doi: 10.1038/nature04733. [DOI] [PubMed] [Google Scholar]
  • 113.Wu G, Gentile L, Fuchikami T, Sutter J, Psathaki K, Esteves TC, et al. Initiation of trophectoderm lineage specification in mouse embryos is independent of Cdx2. Development. 2010;137:4159–69. doi: 10.1242/dev.056630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ortega I, Wong DH, Villanueva JA, Cress AB, Sokalska A, Stanley SD, et al. Effects of resveratrol on growth and function of rat ovarian granulosa cells. Fertil Steril. 2012;98:1563–73. doi: 10.1016/j.fertnstert.2012.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Moraloglu O, Engin-Ustun Y, Tonguc E, Var T, Tapisiz OL, Ergun H, et al. The effect of resveratrol on blood pressure in a rat model of preeclampsia. J Matern Fetal Neonatal Med. 2012;25:845–8. doi: 10.3109/14767058.2011.599081. [DOI] [PubMed] [Google Scholar]
  • 116.Bourque SL, Dolinsky VW, Dyck JR, Davidge ST. Maternal resveratrol treatment during pregnancy improves adverse fetal outcomes in a rat model of severe hypoxia. Placenta. 2012;33:449–52. doi: 10.1016/j.placenta.2012.01.012. [DOI] [PubMed] [Google Scholar]
  • 117.Wong DH, Villanueva JA, Cress AB, Sokalska A, Ortega I, Duleba AJ. Resveratrol inhibits the mevalonate pathway and potentiates the antiproliferative effects of simvastatin in rat theca-interstitial cells. Fertil Steril. 2011;96:1252–8. doi: 10.1016/j.fertnstert.2011.08.010. [DOI] [PubMed] [Google Scholar]
  • 118.Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444:337–42. doi: 10.1038/nature05354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Murase T, Misawa K, Haramizu S, Minegishi Y, Hase T. Nootkatone, a characteristic constituent of grapefruit, stimulates energy metabolism and prevents diet-induced obesity by activating AMPK. Am J Physiol Endocrinol Metab. 2010;299:E266–75. doi: 10.1152/ajpendo.00774.2009. [DOI] [PubMed] [Google Scholar]
  • 120.Han CY, Ki SH, Kim YW, Noh K, Leeda Y, Kang B, et al. Ajoene, a stable garlic by-product, inhibits high fat diet-induced hepatic steatosis and oxidative injury through LKB1-dependent AMPK activation. Antioxid Redox Signal. 2011;14:187–202. doi: 10.1089/ars.2010.3190. [DOI] [PubMed] [Google Scholar]
  • 121.Chinnakannu K, Chen D, Li Y, Wang Z, Dou QP, Reddy GP, et al. Cell cycle-dependent effects of 3,3'-diindolylmethane on proliferation and apoptosis of prostate cancer cells. J Cell Physiol. 2009;219:94–9. doi: 10.1002/jcp.21650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Secor E, Hardie G, K M, Froment P, Louden E, Bonick A et al. AMPK agonists in diet supplements and pharma mediate wide-ranging somatic and reproductive effects. . eCAM 2016;Submitted.
  • 123.Yang L, Sha H, Davisson RL, Qi L. Phenformin activates the unfolded protein response in an AMP-activated protein kinase (AMPK)-dependent manner. J Biol Chem. 2013;288:13631–8. doi: 10.1074/jbc.M113.462762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Sakamoto K, Goransson O, Hardie DG, Alessi DR. Activity of LKB1 and AMPK-related kinases in skeletal muscle: effects of contraction, phenformin, and AICAR. Am J Physiol Endocrinol Metab. 2004;287:E310–7. doi: 10.1152/ajpendo.00074.2004. [DOI] [PubMed] [Google Scholar]
  • 125.Hardie DG. AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function. Genes Dev. 2011;25:1895–908. doi: 10.1101/gad.17420111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hawley SA, Ross FA, Chevtzoff C, Green KA, Evans A, Fogarty S, et al. Use of cells expressing gamma subunit variants to identify diverse mechanisms of AMPK activation. Cell Metab. 2010;11:554–65. doi: 10.1016/j.cmet.2010.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Assisted Reproduction and Genetics are provided here courtesy of Springer Science+Business Media, LLC

RESOURCES