Hostname: page-component-78c5997874-j824f Total loading time: 0 Render date: 2024-10-28T04:47:55.887Z Has data issue: false hasContentIssue false

Does Pten have an impact on oogenesis of PCOS mouse models?

Published online by Cambridge University Press:  19 December 2022

T. Onal
Affiliation:
Manisa Celal Bayar University, Faculty Medicine, Department of Histology and Embryology, Manisa, Turkey
P. Tulay*
Affiliation:
Near East University, Faculty of Medicine, Department of Medical Genetics, Nicosia, Cyprus Near East University, DESAM Research Institute, Nicosia, Cyprus
H.S. Vatansever
Affiliation:
Manisa Celal Bayar University, Faculty Medicine, Department of Histology and Embryology, Manisa, Turkey Near East University, DESAM Research Institute, Nicosia, Cyprus
*
Author for correspondence: Pinar Tulay, Near East University, Faculty of Medicine, Department of Medical Genetics; Near East University, DESAM Research Institute, Nicosia, Cyprus; Near East Boulevard, Nicosia North, Cyprus. E-mail: [email protected]
Rights & Permissions [Opens in a new window]

Summary

Polycystic ovary syndrome (PCOS) is a complex disorder in which the aetiology is still not explained very well. The PI3K/PTEN (phosphatidylinositol 3-kinase/phosphatase and tensin homolog deleted on chromosome 10) pathway is an important pathway that is involved in many mechanisms, including proliferation, growth and motility. PTEN plays a role in granulosa cell proliferation and regulates the differentiation process. The aim of this study was to investigate the expression levels of Pten and Pik3ca in PCOS mouse models with and without any treatment procedures. Three groups of mouse models, PCOS, a PCOS group with clomiphene citrate treatment, and a PCOS group with the combination of clomiphene citrate, metformin and pioglitazone treatment, were established. Ovarian tissues, which were obtained from these groups and a control group with no PCOS, were embedded in paraffin and RNA was extracted. cDNA was synthesized and real-time PCR was conducted to evaluate the expression levels of Pten and Pik3ca. The results of this study showed that both Pten and Pik3ca genes were expressed in the ovarian tissues from the mouse models. Although one-way analysis of variance results showed that Pten was expressed significantly differently in the samples, individual Student’s t-tests did not show any significantly different expression levels in each group. This study is important as it shows the expression patterns of two genes in PCOS mouse models with different treatment strategies, including clomiphene citrate, metformin and pioglitazone. The results of this study formed the basis of research studies and investigations into different genes within the PTEN pathway, as well as other pathways that are under investigation.

Type
Research Article
Copyright
© The Author(s), 2022. Published by Cambridge University Press

Introduction

The mammalian ovaries are composed of follicles at different developmental stages. The first stage of follicular growth is gonadotropin independent, in which the follicle forms the primary and secondary follicle from primordial follicles. The second stage of follicle development is responsive to gonadotropins, in which the pre-antral follicles develop into the early antral follicles. The final stage of follicular development is gonadotropin dependent, in which one follicle is recruited and completes development, resulting in ovulation (McNatty et al., Reference McNatty, Reader, Smith, Heath and Juengel2007). Follicle-stimulating hormone (FSH) has been reported to stimulate early follicle development with granulosa cell proliferation and differentiation. At this stage, the successful development of the oocyte depends on communication with the granulosa cells (Banerjee et al., Reference Banerjee, Banerjee, Saraswat, Bandyopadhyay and Kabir2014). The theca cells have a major role in steroidogenesis. The expression of genes involved in apoptosis, survival as well as genes, such as the tumour suppressor genes, are involved in the regulation of the viability of the granulosa cells and therefore the follicles. There are many pathways involved in the regulation of follicular fate, such as the phosphatidylinositol 3-kinase (PI3K) pathway (Reddy et al., Reference Reddy, Shen, Ren, Boman, Lundin, Ottander, Lindgren, Liu, Sun and Liu2005). The PI3K pathway has been reported to be activated by FSH signalling and is further involved in the regulation of B-cell lymphoma 2 (BCL2) proteins (Alam et al., Reference Alam, Maizels, Park, Ghaey, Feiger, Chandel and Hunzicker-Dunn2004). The PI3K/PTEN (PI3K/phosphatase and tensin homolog deleted on chromosome 10) pathway functions in normal metabolic events, such as cell proliferation, growth and motility. Phophatidylinositol-4,5-bisphosphate (PIP2) phosphorylation is catalyzed by PI3K and enables the conversion into phophatidylinositol-3,4,5-bisphosphate (PIP3). PTEN negatively regulates PI3K enabling the conversion into PIP2 from PIP3. PI3K/PTEN, as well as the AKT signalling pathway, play an important role in the development of primordial follicles (Markholt et al., Reference Markholt, Grøndahl, Ernst, Andersen, Ernst and Lykke-Hartmann2012). One of the underlying mechanisms of premature ovarian failure has been suggested to be through the Pten and PI3K signalling pathways. PTEN has been shown to regulate the proliferation of granulosa cells and, furthermore, to regulate the differentiation process of these cells (Goto et al., Reference Goto, Iwase, Ando, Kurotsuchi, Harata and Kikkawa2007). Previously published studies have reported that deletion of Pten in oocyte-specific patterns led to the overactivation of PI3K signalling, leading to the activation prematurely of the whole group of primordial follicles. This resulted in the depletion of primordial follicles during adulthood, leading to premature ovarian failure (Reddy et al., Reference Reddy, Liu, Adhikari, Jagarlamudi, Rajareddy, Shen, Du, Tang, Hämäläinen, Peng, Lan, Cooney, Huhtaniemi and Liu2008). Although the deletion of Pten at the earlier stages resulted in such a drastic outcome, Pten deletion in oocytes from the primary and more developed follicles did not cause oocyte maturation variations and fertility was not compromised (Jagarlamudi et al., Reference Jagarlamudi, Liu, Adhikari, Reddy, Idahl, Ottander, Lundin and Liu2009). Therefore, it is possible that PI3K/PTEN functions and affects folliculogenesis in a developmental phase pattern. Inhibitors of PTEN were shown to enable primordial follicle development leading to the production of mature oocytes with fertile mice (Li et al., Reference Li, Kawamura, Cheng, Liu, Klein, Liu, Duan and Hsueh2010; Adhikari et al., Reference Adhikari, Gorre, Risal, Zhao, Zhang, Shen and Liu2012).

In this study, we aimed to investigate the expression levels of Pten and Pik3ca in mouse models. The mouse models included a control group with normal morphology and normal parameters of ovaries. The study groups included PCOS, the PCOS group with clomiphene citrate treatment and the PCOS group with clomiphene citrate, metformin and pioglitazone treatment.

Materials and methods

Funding was received by Near East University (SAG-2019-1-038). The PCOS mouse models were established at Manisa Celal Bayar University and ethical approval was obtained by the same university (77.637.435-42). The PCOS models were generated as described previously (Tulay et al., Reference Tulay, Onal and Vatansever2022). Briefly, female prepubertal BALB/c mice were used to generate the models. In total, four groups were obtained. The first group was the control group consisting of non-PCOS mice. For groups 2, 3 and 4, all mice were induced with dehydroepiandrosterone (DHEA) to develop PCOS (Merck Millipore) (6 mg/100 g) dissolved in 0.01 ml of 96% ethanol and mixed with 0.09 ml sesame oil (total volume: 0.1 ml) for injection on 20 days consecutively. Body weight was determined each day and the administration of DHEA was adjusted accordingly. Group 2 was the PCOS control group and the mice did not receive any treatment. The third group was treated with clomiphene citrate (CC; 1.5 mg/kg), dissolved in 0.2 ml saline, which was administrated by gavage for 5 days consecutively. The final group was treated with clomiphene citrate (0.15 mg/100 g), metformin (1.2 mg/100 g) and pioglitazone (0.02 mg/100 g) dissolved in 0.2 ml saline, which was injected by gavage for 5 days consecutively. Blood glucose levels were determined by glucose tolerance test using a glucometer following fasting of the mice for 6 h (Accu-Check, Roche) prior to intraperitoneal 2 g/kg of body weight glucose injection at 15, 60 and 120 min (Kauffman et al., Reference Kauffman, Thackray, Ryan, Tolson, Glidewell-Kenney, Semaan, Poling, Iwata, Breen, Duleba, Stener-Victorin, Shimasaki, Webster and Mellon2015).

Cytological and histochemical analysis

The oestrous cycle was determined by analysis of vaginal smear samples at the beginning and end of the treatment with DHEA. Haematoxylin and eosin staining was used to evaluate the oestrous cycle under a light microscope (Olympus BX43, Tokyo, Japan). The proestrus cycle was determined by cytological analysis of the smear samples and by identifying the nucleated and some cornified epithelial cells. When the majority of the cells were cornified, this was identified as the oestrous cycle. The metestrus cycle was classified when smears consisted of mostly leukocytes and some cornified epithelial cells. When leukocytes with a small number of nucleated epithelial cells were observed, this was classified as the diestrous cycle. The ovarian samples were also evaluated to analyze the follicles in the cortex and stroma of the medulla using a light microscope (Olympus BX-43, Tokyo, Japan).

Gene expression analysis

Total RNA was extracted from ovarian samples fixed in paraffin blocks using the FFPE RNA Purification kit (Norgen, Biotek Corporation). The quality and concentration of the RNA were analyzed using a NanoDrop ND-1000 spectrophotometer (Thermo Scientific, USA). Reverse transcription was conducted using the Transcriptor First Strand cDNA Synthesis Kit (Roche, USA). cDNA was synthesized using random hexamers as well as anchored-oligo(dT)18 primers. Real-time PCR (SYBR Green The LightCycler® 480, USA) was performed to investigate the expression levels of Pten, Pik3 and Pik3ca. The primers were designed using the Primer3 tool to flank two exonic junctions to avoid any DNA amplification (Untergasser et al., Reference Untergasser, Cutcutache, Koressaar, Ye, Faircloth, Remm and Rozen2012) (Table 1). The annealing temperature for all the primers was 55°C. A negative control was used with each run. The Ct values for each gene were obtained using Rotar Gene software (Qiagen) (Table 2). The level of gene expression was normalized against the Actb housekeeping gene. The fold changes were calculated using the comparative ΔΔCq (Tulay et al., Reference Tulay, Naja, Cascales-Roman, Doshi, Serhal and SenGupta2015).

Table 1. Primer sequences. Table shows the forward (F) and reverse (R) sequences of the primers

Table 2. The average Ct values for each gene analysed

Statistical analysis

Statistical analysis was performed to analyze the differences in the expression levels of genes in each mouse model using analysis of variance (ANOVA) test followed by Dunnett’s correction. Two-tailed Student’s t-test was performed to compare the expression level of each gene in the models. GraphPad Prism v8 software was used for all the evaluations.

Results

In this study, PCOS mouse models were successfully generated and different treatment strategies were applied to evaluate the effect on the expression levels of genes involved in the negative feedback mechanism of the PI3K signalling pathway. Overall, it was observed that the ovarian samples obtained from the control group (non-PCOS) preserved follicles were of different sizes as expected. In more detail, follicles at different stages, primordial, primary, secondary and tertiary, presented with expected epithelium and granulosa cells. Furthermore, blood vessels and some loose connective tissue were present in these ovarian samples. The PCOS group presented cysts and follicles of different sizes. In both treatment groups, follicles of different sizes were observed, although groups of cystic follicles were also observed in some places. Irregular oestrus cycles and an increase in weight gain were detected in the DHEA-treated mouse models. Increased levels of basal glucose and abnormalities of glucose tolerance were observed more in the PCOS group compared with the control group. Furthermore, in the PCOS group, immature oocytes within the developing follicles were observed. In these samples, cystic and atretic follicles were also detected. In the larger follicles, the granulosa cells were not as compact as in the control group. Accumulation of lipids was also observed in the granulosa cells. In the treatment groups, the follicles were similar to the control group and the granulosa and theca cells were comparable with those of normal follicles.

The expression levels of two genes, Pten and Pik3ca, were investigated in ovarian tissue samples obtained from the four groups. Gapdh and Actb were used as housekeeping genes and normalization was performed for each analysis against Gapdh and Actb. The results of this study showed that all these genes were expressed in mouse ovaries. The Ct values for each gene are shown in Table 2. The results of the one-way ANOVA showed that the expression levels of Pten were significantly different in the study groups. However, statistical analysis using Student’s t-test for each group individually showed that there was no significant difference.

Discussion

In the present study, the expression levels of Pten and Pik3ca were investigated in PCOS mouse models treated with clomiphene citrate only or a combination of clomiphene citrate, metformin and pioglitazone. The results of this study showed that Pten expression was significantly different in the PCOS mouse models and the treatment groups. Therefore, it is suggested that Pten regulates PCOS development possibly through the PI3K/AKT pathway.

The PI3K/AKT pathway is important in many cellular functions such as the cell cycle, proliferation and cancer. The negative regulatory role of PTEN in the PI3K/AKT pathway has been well established. The roles of PTEN as a tumour suppressor have long been established. However, the PI3K/AKT signalling pathway, as well as the regulation of PTEN through this pathway, have been proposed to be involved in the pathogenesis of PCOS that may be involved in the adjustment of insulin resistance and hyperandrogenism. PTEN has been shown to be associated with insulin concentrations in the follicular fluid (Iwase et al., Reference Iwase, Goto, Harata, Takigawa, Nakahara, Suzuki, Manabe and Kikkawa2009). Expression levels of PTEN have been reported to increase during follicular development. Simultaneously, phospho-AKT has been shown to be reduced, implying a regulatory role for PTEN in granulosa cell development by regulating the PI3K/AKT pathway (Goto et al., Reference Goto, Iwase, Ando, Kurotsuchi, Harata and Kikkawa2007). In the PI3K/AKT pathway, the levels of PIP3 are regulated through the PI3K and PTEN balance. PIP3 is involved in regulating the signalling of many downstream genes, including Akt. Akt has been shown to regulate folliculogenesis (Iwase et al., Reference Iwase, Goto, Harata, Takigawa, Nakahara, Suzuki, Manabe and Kikkawa2009). PTEN has also been suggested to be involved in the development and atresia of primordial follicles (Ouyang et al., Reference Ouyang, Luo, Sun, Huang, Tang, Wu, Zheng and Zheng2013). PTEN has also been shown to be present in granulosa cells (Iwase et al., Reference Iwase, Goto, Harata, Takigawa, Nakahara, Suzuki, Manabe and Kikkawa2009). Furthermore, PTEN was shown to be upregulated in PCOS patients and mouse models, respectively. Therefore, it has been suggested that PTEN regulates the PI3K/AKT signalling pathway and is involved in PCOS development (Gao et al., Reference Gao, Chen, Ji, Ding, Zhang and Yang2021). Furthermore, PTEN expression was shown to be induced with insulin in human granulosa cells leading to reduced Akt phosphorylation. This had a negative effect on the proliferation of granulosa cells. The results of this study showed that Pten was expressed significantly differently in the PCOS and treatment groups. Therefore, it is a possibility that Pten has suppressive effects on the development and function of granulosa cells. The exact mechanism of how PTEN regulates granulosa cell functioning and therefore oogenesis is not clear. Even though some studies have shown decreased expression of PTEN in granulosa cells of PCOS (He et al., Reference He, Liu, Zhao, Liu, Wang and Shi2019), others have reported higher expression of PTEN in the granulosa cells of PCOS patients (Iwase et al., Reference Iwase, Goto, Harata, Takigawa, Nakahara, Suzuki, Manabe and Kikkawa2009). However, it has also been reported that PTEN is downregulated when granulosa cells are proliferating and differentiating in human ovaries and therefore regulating oogenesis through the PI3K pathway (Goto et al., Reference Goto, Iwase, Harata, Takigawa, Suzuki, Manabe and Kikkawa2009). The studies in mice showed that, in the absence of Pten, higher levels of androgens and high numbers of antral follicles with larger ovaries and fertility issues are observed that resemble the PCOS profile in humans (He et al., Reference He, Liu, Zhao, Liu, Wang and Shi2019).

In conclusion, together with previously published studies, this study suggests that Pten is involved in the regulation of PCOS. Treatment strategies can have an effect on the pathogenesis of PCOS by regulating Pten expression. Further studies are ongoing in investigating gene expression levels of other pathways in the pathogenesis of PCOS.

Funding

This research project was funded by the Near East University (SAG-2019-1-038).

Conflict of interest

None to declare.

References

Adhikari, D., Gorre, N., Risal, S., Zhao, Z., Zhang, H., Shen, Y. and Liu, K. (2012). The safe use of a PTEN inhibitor for the activation of dormant mouse primordial follicles and generation of fertilizable eggs. PLOS ONE, 7(6), e39034. doi: 10.1371/journal.pone.0039034 CrossRefGoogle ScholarPubMed
Alam, H., Maizels, E. T., Park, Y., Ghaey, S., Feiger, Z. J., Chandel, N. S. and Hunzicker-Dunn, M. (2004). Follicle-stimulating hormone activation of hypoxia-inducible factor-1 by the phosphatidylinositol 3-kinase/AKT/Ras homolog enriched in brain (Rheb)/mammalian target of rapamycin (mTOR) pathway is necessary for induction of select protein markers of follicular differentiation. Journal of Biological Chemistry, 279(19), 1943119440. doi: 10.1074/jbc.M401235200 CrossRefGoogle ScholarPubMed
Banerjee, S., Banerjee, S., Saraswat, G., Bandyopadhyay, S. A. and Kabir, S. N. (2014). Female reproductive aging is master-planned at the level of ovary. PLOS ONE, 9(5), e96210. doi: 10.1371/journal.pone.0096210 CrossRefGoogle ScholarPubMed
Gao, Y., Chen, J., Ji, R., Ding, J., Zhang, Y. and Yang, J. (2021). USP25 regulates the proliferation and apoptosis of ovarian granulosa cells in polycystic ovary syndrome by modulating the PI3K/AKT pathway via deubiquitinating PTEN. Frontiers in Cell and Developmental Biology, 9, 779718. doi: 10.3389/fcell.2021.779718 CrossRefGoogle ScholarPubMed
Goto, M., Iwase, A., Ando, H., Kurotsuchi, S., Harata, T. and Kikkawa, F. (2007). PTEN and Akt expression during growth of human ovarian follicles. Journal of Assisted Reproduction and Genetics, 24(11), 541546. doi: 10.1007/s10815-007-9156-3 CrossRefGoogle ScholarPubMed
Goto, M., Iwase, A., Harata, T., Takigawa, S., Suzuki, K., Manabe, S. and Kikkawa, F. (2009). IGF1-induced AKT phosphorylation and cell proliferation are suppressed with the increase in PTEN during luteinization in human granulosa cells. Reproduction (Cambridge, England), 137(5), 835842. doi: 10.1530/REP-08-0315 CrossRefGoogle ScholarPubMed
He, T., Liu, Y., Zhao, S., Liu, H., Wang, Z. and Shi, Y. (2019). Comprehensive assessment the expression of core elements related to IGFIR/PI3K pathway in granulosa cells of women with polycystic ovary syndrome. European Journal of Obstetrics, Gynecology, and Reproductive Biology, 233, 134140. doi: 10.1016/j.ejogrb.2018.12.010 CrossRefGoogle ScholarPubMed
Iwase, A., Goto, M., Harata, T., Takigawa, S., Nakahara, T., Suzuki, K., Manabe, S. and Kikkawa, F. (2009). Insulin attenuates the insulin-like growth factor-I (IGF-I)-Akt pathway, not IGF-I-extracellularly regulated kinase pathway, in luteinized granulosa cells with an increase in PTEN. Journal of Clinical Endocrinology and Metabolism, 94(6), 21842191. doi: 10.1210/jc.2008-1948 CrossRefGoogle Scholar
Jagarlamudi, K., Liu, L., Adhikari, D., Reddy, P., Idahl, A., Ottander, U., Lundin, E. and Liu, K. (2009). Oocyte-specific deletion of Pten in mice reveals a stage-specific function of PTEN/PI3K signaling in oocytes in controlling follicular activation. PLOS ONE, 4(7), e6186. doi: 10.1371/journal.pone.0006186.CrossRefGoogle ScholarPubMed
Kauffman, A. S., Thackray, V. G., Ryan, G. E., Tolson, K. P., Glidewell-Kenney, C. A., Semaan, S. J., Poling, M. C., Iwata, N., Breen, K. M., Duleba, A. J., Stener-Victorin, E., Shimasaki, S., Webster, N. J. and Mellon, P. L. (2015). A novel letrozole model recapitulates both the reproductive and metabolic phenotypes of polycystic ovary syndrome in female mice. Biology of Reproduction, 93(3), 69. doi: 10.1095/biolreprod.115.131631 CrossRefGoogle ScholarPubMed
Li, J., Kawamura, K., Cheng, Y., Liu, S., Klein, C., Liu, S., Duan, E. K. and Hsueh, A. J. (2010). Activation of dormant ovarian follicles to generate mature eggs. Proceedings of the National Academy of Sciences of the United States of America, 107(22), 1028010284. doi: 10.1073/pnas.1001198107 CrossRefGoogle ScholarPubMed
Markholt, S., Grøndahl, M. L., Ernst, E. H., Andersen, C. Y., Ernst, E. and Lykke-Hartmann, K. (2012). Global gene analysis of oocytes from early stages in human folliculogenesis shows high expression of novel genes in reproduction. Molecular Human Reproduction, 18(2), 96110. doi: 10.1093/molehr/gar083 CrossRefGoogle ScholarPubMed
McNatty, K. P., Reader, K., Smith, P., Heath, D. A. and Juengel, J. L. (2007). Control of ovarian follicular development to the gonadotrophin-dependent phase: A 2006 perspective. Society of Reproduction and Fertility Supplement, 64, 5568. doi: 10.5661/rdr-vi-55 Google Scholar
Ouyang, J. X., Luo, T., Sun, H. Y., Huang, J., Tang, D. F., Wu, L., Zheng, Y. H. and Zheng, L. P. (2013). RNA interference mediated Pten knock-down inhibit the formation of polycystic ovary. Molecular and Cellular Biochemistry, 380(1–2), 195202. doi: 10.1007/s11010-013-1673-z CrossRefGoogle ScholarPubMed
Reddy, P., Shen, L., Ren, C., Boman, K., Lundin, E., Ottander, U., Lindgren, P., Liu, Y. X., Sun, Q. Y. and Liu, K. (2005). Activation of AKT (PKB) and suppression of FKHRL1 in mouse and rat oocytes by stem cell factor during follicular activation and development. Developmental Biology, 281(2), 160170. doi: 10.1016/j.ydbio.2005.02.013 CrossRefGoogle ScholarPubMed
Reddy, P., Liu, L., Adhikari, D., Jagarlamudi, K., Rajareddy, S., Shen, Y., Du, C., Tang, W., Hämäläinen, T., Peng, S. L., Lan, Z. J., Cooney, A. J., Huhtaniemi, I. and Liu, K. (2008). Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool. Science, 319(5863), 611613. doi: 10.1126/science.1152257 CrossRefGoogle ScholarPubMed
Tulay, P., Naja, R. P., Cascales-Roman, O., Doshi, A., Serhal, P. and SenGupta, S. B. (2015). Investigation of microRNA expression and DNA repair gene transcripts in human oocytes and blastocysts. Journal of Assisted Reproduction and Genetics, 32(12), 17571764. doi: 10.1007/s10815-015-0585-0 CrossRefGoogle ScholarPubMed
Tulay, P., Onal, T. and Vatansever, S. (2022). Molecular regulation of polycystic ovary syndrome: altered gene expression levels in mouse models pretreatment and post-treatment. Zygote (Cambridge, England), 30(3), 352357. doi: 10.1017/S0967199421000769 CrossRefGoogle ScholarPubMed
Untergasser, A., Cutcutache, I., Koressaar, T., Ye, J., Faircloth, B. C., Remm, M. and Rozen, S. G. (2012). Primer3 – New capabilities and interfaces. Nucleic Acids Research, 40(15), e115. doi: 10.1093/nar/gks596 CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Primer sequences. Table shows the forward (F) and reverse (R) sequences of the primers

Figure 1

Table 2. The average Ct values for each gene analysed