Skip to main content
Erschienen in: Reproductive Biology and Endocrinology 1/2018

Open Access 01.12.2018 | Research

Functional role of AKT signaling in bovine early embryonic development: potential link to embryotrophic actions of follistatin

verfasst von: Mohamed Ashry, Sandeep K. Rajput, Joseph K. Folger, Jason G. Knott, Nabil A. Hemeida, Omaima M. Kandil, Refaat S. Ragab, George W. Smith

Erschienen in: Reproductive Biology and Endocrinology | Ausgabe 1/2018

Abstract

Background

TGF-β signaling pathways regulate several crucial processes in female reproduction. AKT is a non-SMAD signaling pathway regulated by TGF-β ligands essential for oocyte maturation and early embryonic development in the mouse, but its regulatory role in bovine early embryonic development is not well established. Previously, we demonstrated a stimulatory role for follistatin (a binding protein for specific members of TGF-β superfamily) in early bovine embryonic development. The objectives of the present studies were to determine the functional role of AKT signaling in bovine early embryonic development and embryotrophic actions of follistatin.

Methods

We used AKT inhibitors III and IV as pharmacological inhibitors of AKT signaling pathway during the first 72 h of in vitro embryo culture. Effects of AKT inhibition on early embryonic development and AKT phosphorylation were investigated in the presence or absence of exogenous follistatin.

Results

Pharmacological inhibition of AKT signaling resulted in a significant reduction in early embryo cleavage, and development to the 8- to 16-cell and blastocyst stages (d7). Treatment with exogenous follistatin increased AKT phosphorylation and rescued the inhibitory effect of AKT inhibitors III and IV on AKT phosphorylation and early embryonic development.

Conclusions

Collectively, results suggest a potential requirement of AKT for bovine early embryonic development, and suggest a potential role for follistatin in regulation of AKT signaling in early bovine embryos.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12958-017-0318-6) contains supplementary material, which is available to authorized users.
Abkürzungen
AKT/PKB
Protein Kinase B
BMPs
Bone Morphogenetic Proteins
BSA
Bovine Serum Albumin
CDK1
Cyclin Dependent Kinase 1
COCs
Cumulus Oocyte Complexes
D
Day
Dpi
Days post insemination
FBS
Fetal Bovine Serum
FSH
Follicle Stimulating Hormone
H
Hour
Hpi
Hour Post Insemination
IGF-1
Insulin Like Growth Factor − 1
Inh
Inhibitor
IVC
In Vitro Culture
IVF
In Vitro Fertilization
IVM
In Vitro Maturation
KSOM
Potassium Simplex Optimization Medium
LH
Luteinizing Hormone
MI
Metaphase I
MII
Metaphase II
pAKT
Phosphorylated AKT
PDE3A
Phosphodiesterase 3A
PVDF
Polyvinylidene Difluoride
Ser
Serine
tAKT
Total AKT
TBST
Tris Buffered Saline
TGF-β
Transforming growth factor β
Thr
Threonine
WB
Western blotting
μM
Micro molar

Background

Poor oocyte quality limits the efficiency of in vitro embryo production in cattle and women [1]. Our previous studies demonstrated a positive association between the transcript abundance of follistatin and oocyte developmental competence using the prepubertal calf and brilliant cresyl blue screening models [2, 3]. We also found that maternally derived follistatin is essential for bovine early embryonic development and exogenous supplementation of follistatin during first 72 h (h) of in vitro embryo culture has a stimulatory effect on early cleavage, embryonic development to 8- to 16-cell and blastocyst stages and blastocyst cell lineage allocation [4]. The functional requirement of follistatin in oocyte and early embryonic development has been described in bovine and other mono-ovulatory species [5].
Follistatin is a transforming growth factor β (TGF-β) superfamily binding protein that may exert its embryotrophic effects through modulation of one or more of the SMAD [6, 7] or non-SMAD [810] signaling pathways. We previously demonstrated that the embryotrophic actions of follistatin on development to 8- to 16-cell and blastocyst stages are linked to the SMAD signaling pathways [11, 12]. However, how follistatin promotes early embryonic development, particularly early cleavage, is still not fully understood.
Follistatin was first identified as a high affinity Activin binding protein [13] and its binding affinity to select bone morphogenetic proteins (BMPs) has also been established [14, 15]. Both BMPs [16] and Activin [17] are major regulators of AKT signaling pathway that promotes cell cycle progression during G2/M transition. Studies have shown that AKT stimulates the transition from metaphase I (MI) to metaphase II (MII) in bovine [18], porcine [19], mouse [20, 21] and Xenopus [22] oocytes by activation of Phosphodiesterase 3A (PDE3A) and cyclin dependent kinase 1 (CDK1) required for resumption and regulation of meiosis. AKT was primarily identified as a serine/threonine specific protein kinase that functions downstream of Phosphatidylinositol 3-kinase (PI3K) [23]. Inhibition of AKT with a synthetic small molecule inhibitor (AKT inhibitor III) resulted in arrest of bovine oocytes at MI stage [18]. AKT plays an important role during entry of one cell mouse embryos into the first mitosis as inhibition of AKT resulted in arrest of cell cycle in G1 and G2 phases [24]. Moreover, inhibition of AKT activity compromised the development of mouse embryos to the blastocyst stage [25, 26]. Considering the role of AKT in cell cycle progression in oocytes and embryos, we hypothesized that AKT signaling is required for bovine early embryonic development and is linked to the embryotrophic actions of follistatin during in vitro embryo culture. To test this hypothesis, we determined the functional role of AKT in bovine early embryonic development and investigated the relationship between AKT signaling and the embryotrophic actions of follistatin. Effects of treatment with the AKT inhibitors III and IV on development of early bovine embryos and AKT phosphorylation were analyzed. Effects of exogenous follistatin supplementation on AKT phosphorylation and developmental potential of AKT inhibitor treated embryos were also investigated.

Methods

All chemicals and reagents used were purchased from Sigma- Aldrich (St. Louis, MO) unless stated otherwise.

Oocyte collection and in vitro embryo production

Bovine oocytes were retrieved from ovaries collected at local abattoir in the state of Michigan. Oocyte aspiration, in vitro maturation (IVM), in vitro fertilization (IVF), and in vitro embryo culture (IVC) were performed as previously described [3, 27]. Briefly, morphologically good quality cumulus oocyte complexes (COCs), which have three or more layers of compact cumulus cell and granular, homogenous cytoplasm, were matured in TCM-199 media [supplemented with 50 μg/ml gentamycin sulfate, 0.2 mM sodium pyruvate, 3.67 nM 17β-estradiol, 15.6 nM bovine FSH (Sioux Biochemical, Sioux Center, IA), 156 nM bovine LH (Sioux Biochemical), and 10% v/v defined fetal bovine serum (FBS) (Hyclone, Logan, UT)] at 38.5 °C in 5% CO2 and humidified air for 24 h. IVF was conducted using motile spermatozoa separated by Percoll gradient technique from frozen thawed bovine semen. The matured COCs were co-incubated with motile spermatozoa (1 × 106 sperm/ml) for 18–20 h in fertilization media FIV (114 mM NaCl, 25 mM NaHCO3, 3.2 mM KCl, 0.34 mM NaH2PO4, 0.183 mM penicillin-G, 16.6 mM sodium lactate, 0.5 mM MgCl26H2O, 2.7 mM CaCl22H2O, 0.2 mM sodium pyruvate, 6 mg/ml BSA, and 1.5 U of heparin) at 38.5 °C in 5% CO2 and humidified air. After IVM and IVF, the cumulus cells were stripped off by vortexing for 6 min. Then, presumptive zygotes were washed and cultured in potassium simplex optimization medium (KSOM; EMD Millipore, Billerica, MA) supplemented with 0.3% fatty acid free bovine serum albumin (BSA) until 72 h post insemination (hpi). Then, 8- to 16-cell stage embryos were separated and cultured in fresh KSOM medium supplemented with 0.3% BSA and 10% FBS under same conditions until d 7.

Effect of AKT inhibitors III & IV on early embryonic development

AKT inhibitor III, known as SH6, is a cell permeable reversible substrate competitive phosphatidylinositol analog that prevents the generation of PIP3 by PIK3 which is required for AKT phosphorylation. AKT inhibitor IV is a cell permeable reversible benzimidazole compound that inhibits AKT phosphorylation/activation by targeting the ATP binding site of a kinase upstream of AKT, but downstream of PI3K [28]. The effects of AKT inhibitors III and IV treatment on early embryonic development were investigated as described in Fig. 1. Presumptive zygotes were cultured in KSOM medium supplemented with 0.3% BSA in the presence of 0, 25, 50 or 75 μM AKT inhibitor III (ALX-270-350 - Enzo Life Sciences, Farmingdale, NY), or 0, 1.5, 2.5 or 3.5 μM AKT inhibitor IV (Sigma, 124,011-1MG) until 72 hpi (n = 25–30 presumptive zygotes/group, n = 4 replicates/inhibitor). Embryos at 8- to 16-cell stage were separated, washed and cultured in fresh KSOM medium supplemented with 0.3% BSA and 10% FBS in the absence of inhibitor until d 7. The effects of AKT inhibition on indices of embryo developmental progression including early cleavage rate determined at 30 hpi, total cleavage rate determined at 48 hpi, percentage of embryos developing to the 8- to 16-cell stage at 72 hpi and percentage of embryos developing to the blastocyst stage at d 7 were recorded.

Effect of AKT inhibitors III & IV on AKT phosphorylation in early bovine embryos

To investigate the effect of AKT inhibitors III & IV treatment on AKT signaling activity in bovine embryo, AKT-Ser473 and AKT-Thr308 phosphorylation levels were analyzed. Presumptive zygotes were cultured in KSOM medium supplemented with 0.3% BSA in the presence of 0, 25, 50 or 75 μM AKT inhibitor III (Fig. 1) [18], or 0, 1.5, 2.5 or 3.5 μM AKT inhibitor IV for 10 h. Then, samples were collected for Western blot analysis of phosphorylated (p)-AKT-Ser473 or pAKT-Thr308, total (t)-AKT and actin (n = 3 replicates/inhibitor/phosphorylation site, n = 20 presumptive zygotes/group). All the doses of AKT inhibitors used in this study were selected based on previously published studies in bovine and mice [18, 19, 21].

Effects of follistatin supplementation on the developmental capacity of AKT inhibitor treated bovine embryos

Experiment was performed as described in Fig. 1. After IVM and IVF, presumptive zygotes were cultured with 0 or 10 ng/ml recombinant human follistatin (R&D Systems, Minneapolis, MN) [4] in the presence or absence of AKT inhibitor III (75 μM) or AKT inhibitor IV (3.5 μM) until 72 hpi (n = 4 replicates/inhibitor, n = 20–30 zygotes/group). Then, 8- to 16-cell stage embryos from each group were separated and cultured in fresh KSOM medium supplemented with 0.3% BSA and 10% FBS without any treatment until d 7. Effects of exogenous follistatin on early and total cleavage, development to 8–16-cell and d7 blastocyst rates were recorded.

Effect of follistatin supplementation on activity of AKT signaling pathway in AKT inhibitor treated early bovine embryos

Effects of follistatin supplementation on AKT signaling activity were investigated by analysis of AKT phosphorylation levels at 10 and 24 h after follistatin supplementation in the presence and absence of AKT inhibitor III or IV (Fig. 1). Presumptive zygotes were cultured with 0 or 10 ng/ml recombinant human follistatin in the presence or absence of AKT inhibitor III (75 μM) or AKT inhibitor IV (3.5 μM) during the initial 72 h of in vitro embryo culture. Samples were collected at 10 h after treatment administration (n = 6 replicates/inhibitor, n = 20 zygotes/group). For the second experiment, follistatin (0 or 10 ng/ml) was supplemented during the same time window and samples collected at 24 h after treatment administration (n = 5 replicates/inhibitor, n = 20 zygotes/group). Samples were subjected to Western blot analysis of pAKT-Ser473, pAKT-Thr308, tAKT and actin.

Western blot analysis

Western blot analysis was performed as described before [3]. Proteins (20 oocytes/embryos per sample) were separated by SDS-PAGE electrophoresis using 4–20% gradient pre-casted gel (Bio-Rad, Hercules, CA) and transferred to a polyvinylidene difluoride (PVDF, Millipore) membrane. After transfer, membranes were blocked with 5% BSA in Tris Buffered Saline with 0.5% Tween 20 (TBST) for 1 h at room temperature before overnight incubation with the appropriate primary antibody diluted in TBST with 5% BSA at 4 °C. The membrane was then washed and incubated with the appropriate HRP-conjugated secondary antibody diluted in TBST with 5% BSA for 1 h at room temperature. Protein signals were detected with SuperSignal West Dura Chemiluminescent Substrate (Thermo Scientific, Waltham, MA) and Images were captured using myECL Imager (Thermo Scientific). ImageJ software was used to measure the band intensities of the target proteins [29]. Relative abundance of each protein was normalized to the total–actin expression in corresponding lane, and phosphorylation level was expressed as phosphorylated (p) AKT/total (t) AKT. Membranes were probed sequentially with primary pAKT-Ser473 rabbit polyclonal antibody [1:1000 (Vol/Vol), Santa Cruz, sc-7985-R] or pAKT-Thr308 rabbit polyclonal antibody [1:1000 (Vol/Vol), Cell Signaling Technology, 9275]. After detection of pAKT membranes were striped with restore Western blot stripping buffer (Thermo Scientific) for 15 min at room temperature and re-probed with tAKT Rabbit polyclonal antibody [1:2000 (Vol/Vol), Santa Cruz, sc-8312]. After detection of tAKT, membranes were stripped and re-probed with actin monoclonal antibody (1:5000 (Vol/Vol) Millipore; MAB1501). HRP-conjugated Anti-Rabbit-IgG (Cell Signaling Technology, 7074), and Anti-Mouse-IgG (Thermo Scientific, A16011) were used as secondary antibodies at a 1:5000 (Vol/Vol) dilution.

Statistical analysis

Differences in phosphorylation levels obtained by Western-blot analysis were analyzed by ANOVA using the general linear models’ procedure of SAS (SAS Institute Inc., Cary, NC). For experiments studying the effects of AKT inhibitors and follistatin supplementation on early embryonic development, data were arcsine transformed prior to analysis by mixed linear models’ analysis procedures. Differences among treatment means were compared using Fisher’s protected least significant difference test. In all cases data are presented as untransformed mean ± standard error (P < 0.05).

Results

AKT inhibition during the initial 72 h of in vitro culture reduced indices of embryonic developmental progression

To elucidate the potential role of AKT in bovine early embryogenesis, effects of AKT inhibitor III and AKT inhibitor IV supplementation (during the initial 72 h of in vitro embryo culture) on different developmental indices (early cleavage, total cleavage, 8- to 16-cell and blastocyst formation rates) were determined. The addition of 25, 50 and 75 μM AKT inhibitor III to the culture medium delayed early cleavage therefore no cleavage was observed at 30 hpi (Fig. 2a). Treatment with AKT inhibitor III significantly reduced the total cleavage rate, percentage of embryos that reached 8- to 16-cell stage at 72 hpi, and d 7 blastocyst rate relative to controls at 75 μM dose (Fig. 2b-d). However, no significant effects on total cleavage were observed in response to both 25 and 50 μM doses compared with control (Fig. 2b, c). Additionally, the 25-mM dose didn’t impact the development to 8-to 16-cell or blastocyst stages (Fig. 2c-d). The addition of AKT inhibitor IV resulted in a dose dependent reduction in early cleavage, development to 8- to 16-cell and blastocyst stage with maximum response observed for 3.5 μM dose which delayed early cleavage and completely blocked development to blastocyst. Total cleavage wasn’t impacted by lower doses of AKT inhibitor IV (1.5, 2.5 μM) whereas the higher dose (3.5 μM) significantly reduced the number of embryos reaching two cells at 48 hpi (Fig. 2e-h). Results demonstrate that inhibition of AKT delayed early embryonic cleavage at 30 hpi, and consequently reduced development to the later endpoints, suggesting a potential requirement of AKT for early embryonic development.

Effects of AKT inhibitors III & IV on AKT phosphorylation in early bovine embryos

Dose response studies were conducted to determine the effects of different concentrations of AKT inhibitor III (25, 50, and 75 μM) or AKT inhibitor IV (1.5, 2.5 and 3.5 μM) on AKT-Ser 473 and Thr308 phosphorylation levels in early embryos after 10 h of treatment. AKT-Thr308 phosphorylation level was significantly reduced in response to high doses of AKT inhibitor III (75 μM) and AKT inhibitor IV (3.5 μM) whereas slight non-significant reduction (P < .09) in phosphorylation level was also observed with lower doses of AKT inhibitor III (25 or 50 μM) or AKT inhibitor IV (1.5 and 2.5 μM) (Fig. 3a, b). On the other hand, AKT-Ser473 phosphorylation level was not impacted by treatment with the 25 μM AKT inhibitor III dose, whereas 50, and 75 μM doses, resulted in a significant reduction after 10 h of supplementation (Additional file 1: Figure S1a). Treatment with AKT inhibitor IV didn’t show any effect on AKT-Ser437 phosphorylation at this time point (Additional file 1: Figure S1b).

Follistatin supplementation rescues the adverse effects of AKT inhibition on early embryo development

Our previous studies established a functional requirement of maternally derived follistatin for bovine early embryogenesis and embryotrophic effects of exogenous follistatin supplementation on early embryo development including enhanced early cleavage, and increased blastocyst formation rate and trophectoderm cell numbers. The AKT signaling pathway is regulated by members of TFG-β superfamily [8]. Hence, we investigated if follistatin supplementation can rescue the negative effects of AKT inhibition on early embryonic development. In the absence of the AKT inhibitors, follistatin supplementation (10 ng/ml) significantly increased the proportion of embryos reaching 2-cell stage at 30 hpi (early cleavage), the proportion of embryos reaching 8- to 16-cell stage at 72 h and d7 blastocyst rates compared with untreated controls. In addition, follistatin supplementation rescued the inhibitory effects of AKT inhibitors on early embryonic development (Fig. 4a-h). Follistatin supplementation was able to rescue the effects of AKT inhibitor III on early cleavage, total cleavage and development to 8- to 16-cell stage to levels similar to controls (Fig. 4a-c), and to partially rescue the effects of AKT inhibitor III on blastocyst development rate (Fig. 4d). Using AKT inhibitor IV with same experimental design, we observed that follistatin supplementation (10 ng/ml) partially rescued the negative effects of AKT inhibitor IV on total cleavage, early cleavage, 8- to 16-cell and blastocysts development rates (Fig. 4e-h). Collectively, results suggest a potential requirement of AKT for bovine early embryonic development, and a possible relationship between the embryotrophic actions of follistatin and the AKT signaling pathway.

Follistatin supplementation modulates the AKT signaling pathway in early bovine embryos

Our results revealed that exogenous follistatin could rescue the negative effects of AKT inhibition on various developmental endpoints in bovine embryos. Therefore, we analyzed the effect of exogenous follistatin supplementation on AKT signaling activity in the presence or absence of AKT inhibitors to determine whether follistatin rescue the effects of AKT inhibition through modulation of AKT signaling. Western blot analysis showed that AKT inhibitor IV treatment resulted in a significant reduction in AKT-Thr308 phosphorylation level in zygotes 10 h post treatment that was rescued by supplementation with exogenous follistatin. However, no effect of follistatin treatment on basal levels of AKT phosphorylation was observed in the absence of inhibitor treatment (Fig. 5a). Similar pattern was observed in response to AKT inhibitor III treatment (Additional file 2: Figure S2a). We further investigated if follistatin supplementation has any effect on basal AKT phosphorylation at a later time point. At 24 h post treatment administration, follistatin supplementation resulted in a significant increase in AKT phosphorylation relative to embryos cultured in the absence of follistatin (Fig. 5b, Additional file 2: Figure S2b).

Discussion

A growing body of evidence supports an important intrinsic role for follistatin in bovine oocyte quality and early embryo developmental progression in vitro [3, 4, 11, 12]. While follistatin is best known for its ability to bind and inhibit activity of select TGF-β superfamily ligands, the intrinsic ligands and signaling pathways linked to trophic actions of follistatin on early embryos are not known. Results of the present study demonstrated that pharmacological inhibition of AKT signaling pathway in early bovine embryos reduced their developmental capacity as assessed by early cleavage, total cleavage and rates of development to 8–16-cell and blastocyst stages. Follistatin supplementation during the first 72 h of in vitro embryo culture rescued the adverse effects of AKT signaling inhibition on embryo development and increased the level of AKT phosphorylation, suggesting a potential role for follistatin in regulation of AKT signaling in early bovine embryos. Results demonstrate follistatin regulation of non-canonical TGF-beta superfamily signaling in bovine embryos and a functional link to developmental progression.
AKT is serine/threonine-specific protein kinase that regulates crucial cellular processes such as glucose metabolism, transcription, cellular growth and proliferation [3032]. Full activation of AKT requires translocation to the plasma membrane and phosphorylation at Thr308 and Ser473 motifs [33]. Previous studies have established a functional role of AKT during oocyte maturation in various species including bovine [18] and in early embryogenesis in mouse and pigs [19, 20, 34]. In bovine embryo, activation of PI3K/AKT pathway by insulin like growth factor − 1 (IGF-1) has been shown to block heat shock induced apoptosis [35]. In addition, pharmacological inhibition of PI3K, upstream kinase of AKT, using LY294002 resulted in significant decrease in total cleavage, 8 cells and blastocyst development rates in bovine [36]. In the present study, we used two different pharmacological inhibitors of AKT activity to investigate its role in early bovine embryo development. Pharmacological inhibition was selected over siRNA mediated knockdown because inhibitors are membrane permeable and can function immediately by specifically targeting AKT activity without affecting its total protein abundance in embryo. As inhibitors can function in reversible manner, this allowed us to inhibit AKT signaling for define period (d1–3) of in vitro embryo culture in this study. Our does dependent study showed that AKT inhibitor III at 75 μm or AKT inhibitor IV at 3.5 μM doses exhibited about 50% reduction in AKT phosphorylation levels and therefore were selected as most effective dose to inhibit AKT signaling. These results were in accordance with the previously published studies in bovine and mice [18, 19, 21]. In addition, higher doses of AKT inhibitor III used in this study has been previously reported to have no nonspecific effect on other kinases downstream to PDK1 [18].
Our results showed that inhibition of AKT during first 3 days of in vitro embryo culture (before zygotic genome activation) resulted in pronounced reduction in multiple indices of early embryonic development. Early embryonic cleavage at 30 hpi was not detected for all tested doses of AKT inhibitor III. Total cleavage, rates of development to 8- to 16-cell and day 7 blastocyst rates were significantly reduced in embryos treated with the highest dose of AKT inhibitor III. We observed similar effects of AKT signaling inhibition on embryo development using AKT inhibitor IV. Data from other species [37, 38] suggest that AKT is implicated in regulation of mitotic cell cycle transition from G2 to M phase through activation of M-phase promoting factor (MPF). Therefore, upon inhibition of AKT, the MPF complex remains inactive and the cells are arrested in G1 or G2 stages of the cell cycle [24, 39]. Reduction of AKT phosphorylation/activity may delay cell cycle progression [40] or compromise the developmental capacity of treated embryos. Therefore, early cleavage at 30 hpi was not detected, and total cleavage, development to 8- to 16-cell and blastocyst stages were reduced compared with untreated embryos. Collectively, these results demonstrated that inhibition of AKT signaling using two different AKT inhibitors similarly reduced the development of early bovine embryos in vitro suggesting an important functional role of AKT signaling in early bovine embryogenesis. However, further mechanistic studies are required to conclusively elucidate how the AKT signaling pathway regulates early embryonic development in bovine.
We previously established a positive association between follistatin and oocyte developmental competence [2, 3]. Supplementation of exogenous follistatin during in vitro embryo culture has embryotropic effects in bovine and non-human primates [4, 41]. Although, follistatin was originally identified as an activin antagonist [13], the embryotropic actions of follistatin are presumably not mediated by inhibition of activin action as exogenous supplementation of activin mimicked the embryotrophic effects of follistatin [4]. As a TGF-β superfamily growth factor binding protein, follistatin may exert its embryotrophic actions by modulating SMAD dependent (SMAD1/5/8 or SMAD2/3) or non-SMAD dependent (AKT, ERK, P38 and JNK) pathways implicated in signaling by this growth factor family [8]. Previous studies in our laboratory demonstrated that, exogenous follistatin supplementation rescued the effects of SMAD2/3 and SMAD4 inhibition/knockdown on early cleavage but not the effects on development to 8- to 16-cell and blastocyst stages, suggesting that the embryotrophic effects of follistatin on early cleavage are SMAD independent [11, 12]. In the current study, the effects of follistatin supplementation on development of AKT inhibitor treated embryos were investigated. Treatment with 10 ng/ml follistatin during the initial 72 h of in vitro embryo culture reversed the inhibitory effects of AKT inhibitor III on early cleavage, total cleavage and development to the 8- to 16-cell stage and partially rescued development to the blastocyst stage. Results also demonstrated that AKT inhibitors significantly reduced the levels of AKT phosphorylation in the absence of follistatin. In addition, follistatin treatment not only reverse the AKT inhibitors effect on phosphorylation level but also increase the AKT phosphorylation when supplemented independently for 24 h. These results suggest that follistatin may be playing a role in returning the AKT phosphorylation to the levels required for rescuing the adverse effect of AKT inhibitors on early cleavage. Collectively, results suggest a potential functional link between follistatin action and AKT signaling linked to its embryotropic actions in bovine embryos. As the effects of other signaling pathways can’t be disregarded, further studies may be required to elucidate the effects of inhibition of multiple signaling pathways on the development of early bovine embryos and to dissect the specific effects of each signaling pathway.
Effects of TGF-β superfamily stimulation on AKT signaling appear to be pleiotropic and cell type dependent. It has been demonstrated that members of TGF-β superfamily activate PI3K pathway through phosphorylation of its downstream effector AKT [4244]. TGF-β can induce a physical interaction between the p85 regulatory subunit of PI3K and the TβRII and TβRI receptors converting phosphatidylinositol-4,5-bisphosphate (PIP2) to phosphatidylinositol-3,4,5-triphosphate (PIP3) and subsequently AKT phosphorylation [4547]. Studies on somatic cell lines indicate that follistatin treatment activates PI3K/AKT signaling pathway [48, 49]. In the present studies, follistatin treatment reversed the inhibitory effects of AKT inhibitor treatment on AKT phosphorylation levels at 10 h post treatment, but a follistatin dependent increase in basal AKT phosphorylation was not observed in the absence of AKT inhibitor treatment. However, follistatin treatment did increase basal AKT phosphorylation levels 24 h post treatment administration. The exact reason for time dependent effects on AKT signaling observed in response to follistatin treatment are not known but may be reflecting time/treatment induced differences in endogenous growth factor milieu and bioaffinity for specific ligands. Results suggest that follistatin plays a role in regulation of AKT signaling in early bovine embryos. However, AKT seems to not be required for the embryotrophic actions of follistatin.

Conclusions

Results of the present study demonstrate that inhibition of AKT signaling reduce the developmental competence of early bovine embryos suggesting a potential requirement of AKT signaling for bovine early embryonic development. Follistatin supplementation rescued the developmental competence of AKT inhibitor treated embryos suggesting that the examined embryotrophic effects of follistatin do not require AKT signaling. However, follistatin supplementation increased the relative abundance of pAKT in AKT inhibitor treated embryos and increased the level of AKT phosphorylation when supplemented without AKT inhibitor, suggesting that follistatin plays a role in regulation of AKT signaling in early bovine embryos. Together, the results reported here, provide additional insights into understanding the regulation of early embryonic development and the mechanism of action of the embryotrophic agent, follistatin, in early bovine embryos.

Acknowledgements

Not applicable.

Funding

This study was supported by the National Institute of Child Health and Human Development of the National Institutes of Health under award number R01HD072972 and by Michigan State University AgBioResearch. MA was supported by the Egyptian Government Joint supervision grant JS2687. The funding agencies had no role neither in study design, data collection, analysis, or interpretation nor in the writing of the report; or in the decision to submit the paper for publication.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analysed during the current study.
Ovaries were collected from local slaughter house in the state of Michigan. Michigan State university institutional animal care and use committee (IACUC) doesn’t require approval for this study.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat Ashry M, Smith GW. Application of embryo transfer using in vitro produced embryos: intrinsic factors affecting efficiency. Cattle Practice. 2015;23:8. Ashry M, Smith GW. Application of embryo transfer using in vitro produced embryos: intrinsic factors affecting efficiency. Cattle Practice. 2015;23:8.
2.
Zurück zum Zitat Patel OV, Bettegowda A, Ireland JJ, Coussens PM, Lonergan P, Smith GW. Functional genomics studies of oocyte competence: evidence that reduced transcript abundance for follistatin is associated with poor developmental competence of bovine oocytes. Reproduction. 2007;133:95–106.CrossRefPubMed Patel OV, Bettegowda A, Ireland JJ, Coussens PM, Lonergan P, Smith GW. Functional genomics studies of oocyte competence: evidence that reduced transcript abundance for follistatin is associated with poor developmental competence of bovine oocytes. Reproduction. 2007;133:95–106.CrossRefPubMed
3.
Zurück zum Zitat Ashry M, Lee K, Mondal M, Datta TK, Folger JK, Rajput SK, Zhang K, Hemeida NA, Smith GW. Expression of TGFbeta superfamily components and other markers of oocyte quality in oocytes selected by brilliant cresyl blue staining: relevance to early embryonic development. Mol Reprod Dev. 2015;82:251–64.CrossRefPubMedPubMedCentral Ashry M, Lee K, Mondal M, Datta TK, Folger JK, Rajput SK, Zhang K, Hemeida NA, Smith GW. Expression of TGFbeta superfamily components and other markers of oocyte quality in oocytes selected by brilliant cresyl blue staining: relevance to early embryonic development. Mol Reprod Dev. 2015;82:251–64.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Lee KB, Bettegowda A, Wee G, Ireland JJ, Smith GW. Molecular determinants of oocyte competence: potential functional role for maternal (oocyte-derived) follistatin in promoting bovine early embryogenesis. Endocrinology. 2009;150:2463–71.CrossRefPubMed Lee KB, Bettegowda A, Wee G, Ireland JJ, Smith GW. Molecular determinants of oocyte competence: potential functional role for maternal (oocyte-derived) follistatin in promoting bovine early embryogenesis. Endocrinology. 2009;150:2463–71.CrossRefPubMed
5.
Zurück zum Zitat Rajput SK, Lee K, Zhenhua G, Di L, Folger JK, Smith GW. Embryotropic actions of follistatin: paracrine and autocrine mediators of oocyte competence and embryo developmental progression. Reprod Fertil Dev. 2013;26:37–47.CrossRefPubMedPubMedCentral Rajput SK, Lee K, Zhenhua G, Di L, Folger JK, Smith GW. Embryotropic actions of follistatin: paracrine and autocrine mediators of oocyte competence and embryo developmental progression. Reprod Fertil Dev. 2013;26:37–47.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Moustakas A, Souchelnytskyi S, Heldin CH. Smad regulation in TGF-beta signal transduction. J Cell Sci. 2001;114:4359–69.PubMed Moustakas A, Souchelnytskyi S, Heldin CH. Smad regulation in TGF-beta signal transduction. J Cell Sci. 2001;114:4359–69.PubMed
7.
Zurück zum Zitat Attisano L, Wrana JL. Signal transduction by the TGF-beta superfamily. Science. 2002;296:1646–7.CrossRefPubMed Attisano L, Wrana JL. Signal transduction by the TGF-beta superfamily. Science. 2002;296:1646–7.CrossRefPubMed
9.
10.
Zurück zum Zitat Ikushima H, Miyazono K. TGFbeta signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–24.CrossRefPubMed Ikushima H, Miyazono K. TGFbeta signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–24.CrossRefPubMed
11.
Zurück zum Zitat Zhang K, Rajput SK, Lee KB, et al. Evidence supporting a role for SMAD2/3 in bovine early embryonic development: potential implications for Embryotropic actions of Follistatin. Biol Reprod. 2015;93(4):86.CrossRefPubMedPubMedCentral Zhang K, Rajput SK, Lee KB, et al. Evidence supporting a role for SMAD2/3 in bovine early embryonic development: potential implications for Embryotropic actions of Follistatin. Biol Reprod. 2015;93(4):86.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Lee KB, Zhang K, Folger JK, Knott JG, Smith GW. Evidence supporting a functional requirement of SMAD4 for bovine preimplantation embryonic development: a potential link to embryotrophic actions of follistatin. Biol Reprod. 2014;91:62.PubMedPubMedCentral Lee KB, Zhang K, Folger JK, Knott JG, Smith GW. Evidence supporting a functional requirement of SMAD4 for bovine preimplantation embryonic development: a potential link to embryotrophic actions of follistatin. Biol Reprod. 2014;91:62.PubMedPubMedCentral
13.
Zurück zum Zitat Nakamura T, Takio K, Eto Y, Shibai H, Titani K, Sugino H. Activin-binding protein from rat ovary is follistatin. Science. 1990;247:836–8.CrossRefPubMed Nakamura T, Takio K, Eto Y, Shibai H, Titani K, Sugino H. Activin-binding protein from rat ovary is follistatin. Science. 1990;247:836–8.CrossRefPubMed
14.
Zurück zum Zitat Iemura S, Yamamoto TS, Takagi C, Uchiyama H, Natsume T, Shimasaki S, Sugino H, Ueno N. Direct binding of follistatin to a complex of bone-morphogenetic protein and its receptor inhibits ventral and epidermal cell fates in early Xenopus embryo. Proc Natl Acad Sci U S A. 1998;95:9337–42.CrossRefPubMedPubMedCentral Iemura S, Yamamoto TS, Takagi C, Uchiyama H, Natsume T, Shimasaki S, Sugino H, Ueno N. Direct binding of follistatin to a complex of bone-morphogenetic protein and its receptor inhibits ventral and epidermal cell fates in early Xenopus embryo. Proc Natl Acad Sci U S A. 1998;95:9337–42.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Lin SY, Morrison JR, Phillips DJ, de Kretser DM. Regulation of ovarian function by the TGF-beta superfamily and follistatin. Reproduction. 2003;126:133–48.CrossRefPubMed Lin SY, Morrison JR, Phillips DJ, de Kretser DM. Regulation of ovarian function by the TGF-beta superfamily and follistatin. Reproduction. 2003;126:133–48.CrossRefPubMed
16.
Zurück zum Zitat Ghosh-Choudhury N, Abboud SL, Nishimura R, Celeste A, Mahimainathan L, Choudhury GG. Requirement of BMP-2-induced phosphatidylinositol 3-kinase and Akt serine/threonine kinase in osteoblast differentiation and Smad-dependent BMP-2 gene transcription. J Biol Chem. 2002;277:33361–8.CrossRefPubMed Ghosh-Choudhury N, Abboud SL, Nishimura R, Celeste A, Mahimainathan L, Choudhury GG. Requirement of BMP-2-induced phosphatidylinositol 3-kinase and Akt serine/threonine kinase in osteoblast differentiation and Smad-dependent BMP-2 gene transcription. J Biol Chem. 2002;277:33361–8.CrossRefPubMed
17.
Zurück zum Zitat Do TV, Kubba LA, Antenos M, Rademaker AW, Sturgis CD, Woodruff TK. The role of activin a and Akt/GSK signaling in ovarian tumor biology. Endocrinology. 2008;149:3809–16.CrossRefPubMedPubMedCentral Do TV, Kubba LA, Antenos M, Rademaker AW, Sturgis CD, Woodruff TK. The role of activin a and Akt/GSK signaling in ovarian tumor biology. Endocrinology. 2008;149:3809–16.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Tomek W, Smiljakovic T. Activation of Akt (protein kinase B) stimulates metaphase I to metaphase II transition in bovine oocytes. Reproduction. 2005;130:423–30.CrossRefPubMed Tomek W, Smiljakovic T. Activation of Akt (protein kinase B) stimulates metaphase I to metaphase II transition in bovine oocytes. Reproduction. 2005;130:423–30.CrossRefPubMed
19.
Zurück zum Zitat Kalous J, Kubelka M, Solc P, Susor A, Motlik J. AKT (protein kinase B) is implicated in meiotic maturation of porcine oocytes. Reproduction. 2009;138:645–54.CrossRefPubMed Kalous J, Kubelka M, Solc P, Susor A, Motlik J. AKT (protein kinase B) is implicated in meiotic maturation of porcine oocytes. Reproduction. 2009;138:645–54.CrossRefPubMed
20.
Zurück zum Zitat Kalous J, Solc P, Baran V, Kubelka M, Schultz RM, Motlik J. PKB/AKT is involved in resumption of meiosis in mouse oocytes. Biol Cell. 2006;98:111–23.CrossRefPubMed Kalous J, Solc P, Baran V, Kubelka M, Schultz RM, Motlik J. PKB/AKT is involved in resumption of meiosis in mouse oocytes. Biol Cell. 2006;98:111–23.CrossRefPubMed
21.
Zurück zum Zitat Hoshino Y, Sato E. Protein kinase B (PKB/Akt) is required for the completion of meiosis in mouse oocytes. Dev Biol. 2008;314:215–23.CrossRefPubMed Hoshino Y, Sato E. Protein kinase B (PKB/Akt) is required for the completion of meiosis in mouse oocytes. Dev Biol. 2008;314:215–23.CrossRefPubMed
22.
Zurück zum Zitat Andersen CB, Roth RA, Conti M. Protein kinase B/Akt induces resumption of meiosis in Xenopus oocytes. J Biol Chem. 1998;273:18705–8.CrossRefPubMed Andersen CB, Roth RA, Conti M. Protein kinase B/Akt induces resumption of meiosis in Xenopus oocytes. J Biol Chem. 1998;273:18705–8.CrossRefPubMed
23.
Zurück zum Zitat Bellacosa A, Testa JR, Staal SP, Tsichlis PN. A retroviral oncogene, akt, encoding a serine-threonine kinase containing an SH2-like region. Science. 1991;254:274–7.CrossRefPubMed Bellacosa A, Testa JR, Staal SP, Tsichlis PN. A retroviral oncogene, akt, encoding a serine-threonine kinase containing an SH2-like region. Science. 1991;254:274–7.CrossRefPubMed
24.
Zurück zum Zitat Baran V, Fabian D, Rehak P. Akt/PKB plays role of apoptosis relay on entry into first mitosis of mouse embryo. Zygote. 2013;21:406–16.CrossRefPubMed Baran V, Fabian D, Rehak P. Akt/PKB plays role of apoptosis relay on entry into first mitosis of mouse embryo. Zygote. 2013;21:406–16.CrossRefPubMed
25.
Zurück zum Zitat Li Y, Chandrakanthan V, Day ML, O'Neill C. Direct evidence for the action of phosphatidylinositol (3,4,5)-trisphosphate-mediated signal transduction in the 2-cell mouse embryo. Biol Reprod. 2007;77:813–21.CrossRefPubMed Li Y, Chandrakanthan V, Day ML, O'Neill C. Direct evidence for the action of phosphatidylinositol (3,4,5)-trisphosphate-mediated signal transduction in the 2-cell mouse embryo. Biol Reprod. 2007;77:813–21.CrossRefPubMed
26.
Zurück zum Zitat Riley JK, Carayannopoulos MO, Wyman AH, Chi M, Ratajczak CK, Moley KH. The PI3K/Akt pathway is present and functional in the preimplantation mouse embryo. Dev Biol. 2005;284:377–86.CrossRefPubMed Riley JK, Carayannopoulos MO, Wyman AH, Chi M, Ratajczak CK, Moley KH. The PI3K/Akt pathway is present and functional in the preimplantation mouse embryo. Dev Biol. 2005;284:377–86.CrossRefPubMed
27.
Zurück zum Zitat Bettegowda A, Patel OV, Ireland JJ, Smith GW. Quantitative analysis of messenger RNA abundance for ribosomal protein L-15, cyclophilin-a, phosphoglycerokinase, beta-glucuronidase, glyceraldehyde 3-phosphate dehydrogenase, beta-actin, and histone H2A during bovine oocyte maturation and early embryogenesis in vitro. Mol Reprod Dev. 2006;73:267–78.CrossRefPubMed Bettegowda A, Patel OV, Ireland JJ, Smith GW. Quantitative analysis of messenger RNA abundance for ribosomal protein L-15, cyclophilin-a, phosphoglycerokinase, beta-glucuronidase, glyceraldehyde 3-phosphate dehydrogenase, beta-actin, and histone H2A during bovine oocyte maturation and early embryogenesis in vitro. Mol Reprod Dev. 2006;73:267–78.CrossRefPubMed
28.
29.
Zurück zum Zitat Schneider CA, Rasband WS, Eliceiri KW. NIH image to ImageJ: 25 years of image analysis. Nat Meth. 2012;9:671–5.CrossRef Schneider CA, Rasband WS, Eliceiri KW. NIH image to ImageJ: 25 years of image analysis. Nat Meth. 2012;9:671–5.CrossRef
30.
Zurück zum Zitat Kandel ES, Skeen J, Majewski N, Di Cristofano A, Pandolfi PP, Feliciano CS, Gartel A, Hay N. Activation of Akt/protein kinase B overcomes a G(2)/m cell cycle checkpoint induced by DNA damage. Mol Cell Biol. 2002;22:7831–41.CrossRefPubMedPubMedCentral Kandel ES, Skeen J, Majewski N, Di Cristofano A, Pandolfi PP, Feliciano CS, Gartel A, Hay N. Activation of Akt/protein kinase B overcomes a G(2)/m cell cycle checkpoint induced by DNA damage. Mol Cell Biol. 2002;22:7831–41.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Song G, Ouyang G, Bao S. The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med. 2005;9:59–71.CrossRefPubMed Song G, Ouyang G, Bao S. The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med. 2005;9:59–71.CrossRefPubMed
32.
Zurück zum Zitat Kandel ES, Hay N. The regulation and activities of the multifunctional serine/Threonine Kinase Akt/PKB. Exp Cell Res. 1999;253:210–29.CrossRefPubMed Kandel ES, Hay N. The regulation and activities of the multifunctional serine/Threonine Kinase Akt/PKB. Exp Cell Res. 1999;253:210–29.CrossRefPubMed
33.
Zurück zum Zitat Downward J. Mechanisms and consequences of activation of protein kinase B/Akt. Curr Opin Cell Biol. 1998;10:262–7.CrossRefPubMed Downward J. Mechanisms and consequences of activation of protein kinase B/Akt. Curr Opin Cell Biol. 1998;10:262–7.CrossRefPubMed
34.
Zurück zum Zitat Feng C, Yu A, Liu Y, Zhang J, Zong Z, Su W, Zhang Z, Yu D, Sun QY, Yu B. Involvement of protein kinase B/AKT in early development of mouse fertilized eggs. Biol Reprod. 2007;77:560–8.CrossRefPubMed Feng C, Yu A, Liu Y, Zhang J, Zong Z, Su W, Zhang Z, Yu D, Sun QY, Yu B. Involvement of protein kinase B/AKT in early development of mouse fertilized eggs. Biol Reprod. 2007;77:560–8.CrossRefPubMed
35.
Zurück zum Zitat Jousan FD, Oliveira LJ, Hansen PJ. Short-term culture of in vitro produced bovine preimplantation embryos with insulin-like growth factor-i prevents heat shock-induced apoptosis through activation of the Phosphatidylinositol 3-Kinase/Akt pathway. Mol Reprod Dev. 2008;75:681–8.CrossRefPubMed Jousan FD, Oliveira LJ, Hansen PJ. Short-term culture of in vitro produced bovine preimplantation embryos with insulin-like growth factor-i prevents heat shock-induced apoptosis through activation of the Phosphatidylinositol 3-Kinase/Akt pathway. Mol Reprod Dev. 2008;75:681–8.CrossRefPubMed
36.
Zurück zum Zitat Aparicio IM, Garcia-Herreros M, Fair T, Lonergan P. Identification and regulation of glycogen synthase kinase-3 during bovine embryo development. Reproduction. 2010;140:83–92.CrossRefPubMed Aparicio IM, Garcia-Herreros M, Fair T, Lonergan P. Identification and regulation of glycogen synthase kinase-3 during bovine embryo development. Reproduction. 2010;140:83–92.CrossRefPubMed
37.
Zurück zum Zitat Katayama K, Fujita N, Tsuruo T. Akt/protein kinase B-dependent phosphorylation and inactivation of WEE1Hu promote cell cycle progression at G2/M transition. Mol Cell Biol. 2005;25:5725–37.CrossRefPubMedPubMedCentral Katayama K, Fujita N, Tsuruo T. Akt/protein kinase B-dependent phosphorylation and inactivation of WEE1Hu promote cell cycle progression at G2/M transition. Mol Cell Biol. 2005;25:5725–37.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Okumura E, Fukuhara T, Yoshida H, Hanada Si S, Kozutsumi R, Mori M, Tachibana K, Kishimoto T. Akt inhibits Myt1 in the signalling pathway that leads to meiotic G2/M-phase transition. Nat Cell Biol. 2002;4:111–6.CrossRefPubMed Okumura E, Fukuhara T, Yoshida H, Hanada Si S, Kozutsumi R, Mori M, Tachibana K, Kishimoto T. Akt inhibits Myt1 in the signalling pathway that leads to meiotic G2/M-phase transition. Nat Cell Biol. 2002;4:111–6.CrossRefPubMed
39.
Zurück zum Zitat Roberts EC, Shapiro PS, Nahreini TS, Pages G, Pouyssegur J, Ahn NG. Distinct cell cycle timing requirements for extracellular signal-regulated kinase and phosphoinositide 3-kinase signaling pathways in somatic cell mitosis. Mol Cell Biol. 2002;22:7226–41.CrossRefPubMedPubMedCentral Roberts EC, Shapiro PS, Nahreini TS, Pages G, Pouyssegur J, Ahn NG. Distinct cell cycle timing requirements for extracellular signal-regulated kinase and phosphoinositide 3-kinase signaling pathways in somatic cell mitosis. Mol Cell Biol. 2002;22:7226–41.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Ornelas IM, Silva TM, Fragel-Madeira L, Ventura ALM. Inhibition of PI3K/Akt pathway impairs G2/M transition of cell cycle in late developing progenitors of the avian embryo retina. PLoS One. 2013;8:e53517.CrossRefPubMedPubMedCentral Ornelas IM, Silva TM, Fragel-Madeira L, Ventura ALM. Inhibition of PI3K/Akt pathway impairs G2/M transition of cell cycle in late developing progenitors of the avian embryo retina. PLoS One. 2013;8:e53517.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat VandeVoort CA, Mtango NR, Lee YS, Smith GW, Latham KE. Differential effects of follistatin on nonhuman primate oocyte maturation and pre-implantation embryo development in vitro. Biol Reprod. 2009;81:1139–46.CrossRefPubMedPubMedCentral VandeVoort CA, Mtango NR, Lee YS, Smith GW, Latham KE. Differential effects of follistatin on nonhuman primate oocyte maturation and pre-implantation embryo development in vitro. Biol Reprod. 2009;81:1139–46.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL. Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem. 2000;275:36803–10.CrossRefPubMed Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL. Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem. 2000;275:36803–10.CrossRefPubMed
43.
Zurück zum Zitat Lamouille S, Derynck R. Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 2007;178:437–51.CrossRefPubMedPubMedCentral Lamouille S, Derynck R. Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 2007;178:437–51.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Yi JY, Shin I, Arteaga CL. Type I transforming growth factor beta receptor binds to and activates phosphatidylinositol 3-kinase. J Biol Chem. 2005;280:10870–6.CrossRefPubMed Yi JY, Shin I, Arteaga CL. Type I transforming growth factor beta receptor binds to and activates phosphatidylinositol 3-kinase. J Biol Chem. 2005;280:10870–6.CrossRefPubMed
46.
48.
Zurück zum Zitat Winbanks CE, Weeks KL, Thomson RE, Sepulveda PV, Beyer C, Qian H, Chen JL, Allen JM, Lancaster GI, Febbraio MA, et al. Follistatin-mediated skeletal muscle hypertrophy is regulated by Smad3 and mTOR independently of myostatin. J Cell Biol. 2012;197:997–1008.CrossRefPubMedPubMedCentral Winbanks CE, Weeks KL, Thomson RE, Sepulveda PV, Beyer C, Qian H, Chen JL, Allen JM, Lancaster GI, Febbraio MA, et al. Follistatin-mediated skeletal muscle hypertrophy is regulated by Smad3 and mTOR independently of myostatin. J Cell Biol. 2012;197:997–1008.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Li X, Liu H, Wang H, Sun L, Ding F, Sun W, Han C, Wang J. Follistatin could promote the proliferation of duck primary myoblasts by activating PI3K/Akt/mTOR signalling. Biosci Rep. 2014;34:e00143.PubMedPubMedCentral Li X, Liu H, Wang H, Sun L, Ding F, Sun W, Han C, Wang J. Follistatin could promote the proliferation of duck primary myoblasts by activating PI3K/Akt/mTOR signalling. Biosci Rep. 2014;34:e00143.PubMedPubMedCentral
Metadaten
Titel
Functional role of AKT signaling in bovine early embryonic development: potential link to embryotrophic actions of follistatin
verfasst von
Mohamed Ashry
Sandeep K. Rajput
Joseph K. Folger
Jason G. Knott
Nabil A. Hemeida
Omaima M. Kandil
Refaat S. Ragab
George W. Smith
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Reproductive Biology and Endocrinology / Ausgabe 1/2018
Elektronische ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-017-0318-6

Weitere Artikel der Ausgabe 1/2018

Reproductive Biology and Endocrinology 1/2018 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.