scholarly journals Awakening the oocyte: controlling primordial follicle development

Reproduction ◽  
2009 ◽  
Vol 137 (1) ◽  
pp. 1-11 ◽  
Author(s):  
Eileen A McLaughlin ◽  
Skye C McIver

Oocytes are sequestered in primordial follicles before birth and remain quiescent in the ovary, often for decades, until recruited into the growing pool throughout the reproductive years. Therefore, activation of follicle growth is a major biological checkpoint that controls female reproductive potential. However, we are only just beginning to elucidate the cellular mechanisms required for either maintenance of the quiescent primordial follicle pool or initiation of follicle growth. Understanding the intracellular signalling systems that control oocyte maintenance and activation has significant implications for improving female reproductive productivity and longevity in mammals, and has application in domestic animal husbandry, feral animal population control and infertility in women.

2020 ◽  
Vol 21 (9) ◽  
pp. 3120
Author(s):  
Sook Young Yoon ◽  
Ran Kim ◽  
Hyunmee Jang ◽  
Dong Hyuk Shin ◽  
Jin Il Lee ◽  
...  

Peroxisome proliferator-activated receptor gamma (PPARγ) is known as a regulator of cellular functions, including adipogenesis and immune cell activation. The objectives of this study were to investigate the expression of PPARγ and identify the mechanism of primordial follicle activation via PPARγ modulators in mouse ovaries. We first measured the gene expression of PPARγ and determined its relationship with phosphatase and tensin homolog (PTEN), protein kinase B (AKT1), and forkhead box O3a (FOXO3a) expression in neonatal mouse ovaries. We then incubated neonatal mouse ovaries with PPARγ modulators, including rosiglitazone (a synthetic agonist of PPARγ), GW9662 (a synthetic antagonist of PPARγ), and cyclic phosphatidic acid (cPA, a physiological inhibitor of PPARγ), followed by transplantation into adult ovariectomized mice. After the maturation of the transplanted ovaries, primordial follicle growth activation, follicle growth, and embryonic development were evaluated. Finally, the delivery of live pups after embryo transfer into recipient mice was assessed. While PPARγ was expressed in ovaries from mice of all ages, its levels were significantly increased in ovaries from 20-day-old mice. In GW9662-treated ovaries in vitro, PTEN levels were decreased, AKT was activated, and FOXO3a was excluded from the nuclei of primordial follicles. After 1 month, cPA-pretreated, transplanted ovaries produced the highest numbers of oocytes and polar bodies, exhibited the most advanced embryonic development, and had the greatest blastocyst formation rate compared to the rosiglitazone- and GW9662-pretreated groups. Additionally, the successful delivery of live pups after embryo transfer into the recipient mice transplanted with cPA-pretreated ovaries was confirmed. Our study demonstrates that PPARγ participates in primordial follicle activation and development, possibly mediated in part by the PI3K/AKT signaling pathway. Although more studies are required, adapting these findings for the activation of human primordial follicles may lead to treatments for infertility that originates from poor ovarian reserves.


1994 ◽  
Vol 13 (1) ◽  
pp. 1-9 ◽  
Author(s):  
R Braw-Tal ◽  
D J Tisdall ◽  
N L Hudson ◽  
P Smith ◽  
K P McNatty

ABSTRACT The aim of this study was to investigate the sites of follistatin and α and βA inhibin mRNA expression in the ovaries of female sheep fetuses at 90, 100, 120 and 135 days of gestation (term=day 147). At 90 and 100 days primordial follicles were formed, followed by the appearance of primary follicles at 100 days of gestation. At days 120 and 135, primordial, primary and preantral (i.e. secondary) follicles were present in the ovaries, but antral (i.e. tertiary) follicles were not observed at any of these gestational ages. Two Booroola genotypes were studied: homozygous carriers (BB) and non-carriers (++) of the fecundity gene (FecB). Irrespective of genotype no specific hybridization of the α and βA inhibin riboprobes was detected in any ovarian cells at days 90, 100, 120 or 135 of gestation. In control mature ovaries, on the other hand, strong hybridization in the granulosa cells of antral follicles was observed. In contrast to α and βA inhibin, follistatin antisense (but not sense) riboprobes hybridized specifically to the granulosa cells of preantral follicles with two or more layers of cells at days 120 and 135 of gestation. Moreover, hybridization was also evident in the cells of the ovarian rete at days 120 and 135, but not at 90 or 100 days. No follistatin mRNA expression was observed in the granulosa cells of primordial or primary follicles or in any other ovarian cell type at any of the gestational ages examined. No FecB-specific differences in follistatin expression were noted with respect to stage of preantral follicular development and there were no obvious differences in the intensity of expression. These results show that follistatin mRNA is expressed specifically in the granulosa cells and intraovarian rete. Expression of follistatin in rete cells was coincident with the increasing numbers of growing follicles within the fetal ovary, indicating that rete cell function may have a role in the ontogeny of early follicular growth. Our results suggest that follistatin and α and βA inhibin may not be important for the initiation of follicle growth in the sheep ovary, since these genes are not expressed during the transformation of a primordial follicle to a primary structure. However, the evidence for follistatin mRNA expression in the ovine fetal ovary implies that this hormone is likely to play a role during the early stages of follicle growth.


Reproduction ◽  
2018 ◽  
Author(s):  
John J Peluso ◽  
Xiufang Liu ◽  
Tracy Uliasz ◽  
Cindy A. Pru ◽  
Nicole C. Kelp ◽  
...  

To determine whether conditional depletion of Progesterone Receptor Membrane Component (PGRMC) 1 and PGRMC2 affected ovarian follicle development, follicle distribution was assessed in ovaries of young (≈ 3 month-old) and middle-aged (≈6 month-old) control (Pgrmc1/2fl/fl) and double conditional PGRMC1/2 knockout (Pgrmc1/2d/d) mice. This study revealed that the distribution of primary, preantral and antral follicles was not altered in Pgrmc1/2d/d mice, regardless of the age. Although the number of primordial follicles was similar at ≈ 3 months of age, their numbers were reduced by ≈ 80% in 6-month old Pgrmc1/2d/d mice compared to age-matched Pgrmc1/2fl/fl mice. The Pgrmc1/2d/d mice were generated using Pgr-cre mice, so ablation of Pgrmc1 and Pgrmc2 in the ovary was restricted to peri-ovulatory follicles and subsequent corpora lutea (CL). In addition, the vascularization of CL was attenuated in Pgrmc1/2d/d mice, although mRNA levels of Vascular Endothelial Growth Factor A (Vegfa) were elevated. Moreover, depletion of Pgrmc1 and Pgrmc2 altered the gene expression profile in the non-luteal component of the ovary such that Vegfa expression, a stimulator of primordial follicle growth, was elevated; Kit Ligand expression, another stimulator of primordial follicle growth, was suppressed and Anti-Mullerian Hormone, an inhibitor of primordial follicle growth, was enhanced compared to Pgrmc1/2fl/fl mice. These data reveal that luteal cell depletion of Pgrmc1 and 2 alters the expression of growth factors within the non-luteal component of the ovary which could account for the premature demise of the adult population of primordial follicles.


2013 ◽  
Vol 25 (1) ◽  
pp. 243
Author(s):  
R. L. Krisher ◽  
M. Paczkowski ◽  
K. Maruniak ◽  
W. B. Schoolcraft

Activation and growth of dormant follicles completely in vitro, with the production of competent oocytes, would provide not only the opportunity to investigate control mechanisms of follicle growth, but also an instrument by which fertility might be preserved in women with cancer, premature ovarian failure, or advanced maternal age. The objective of this study was to assess the physical culture parameters, as well as the addition of an antibody to the anti-angiogenic isoform of VEGFA (VEGFAxxxb), for the ability to support activation of primordial follicles and subsequent functional follicular growth in vitro. The phosphatase and tensin homologue (PTEN) inhibitor, bpV(pic), was used to activate growth of primordial follicles in neonatal Day 3 mouse ovaries. BpV(pic) (100 µM) was used either alone, or in combination with FSH (0.3 IU mL–1), IGF-1 (10 ng mL–1), EGF (10 ng mL–1), and ITS (insulin-transferrin-selenium; 1x). Ovaries were treated for 24 h and then cultured for an additional 12 to 20 days, either suspended on a membrane or encapsulated in an alginate bead, with or without VEGFAxxxb during culture. Oestradiol (E2) production and expression of Ahr, Amh, Bmp15, Cyp19, and Esr1, genes known to be involved in follicular growth, were evaluated to assess growth and functionality of follicles. The E2 data were analysed with ANOVA; the qPCR data were analysed using the non-parametric Mann–Whitney U t-test (significance at P < 0.05). Forty-eight hours after treatment with bpV(pic), alone or with FSH, IGF-1, EGF, and ITS, resulted in an upregulation of Amh and Esr1, and a tendency for upregulation of Ahr compared with untreated control ovaries. There were no differences in gene expression between the 2 bpV(pic) treatments. Following treatment and in vitro culture, expression of Ahr, Amh, and Bmp15 was upregulated in ovaries treated and cultured in a bead with VEGFAxxxb compared with ovaries treated and cultured in a bead without VEGFAxxxb, and ovaries treated on a membrane and cultured in a bead with VEGFAxxxb. Although ovaries treated on a membrane with bpV(pic) produced more E2 than untreated ovaries (5.1 ± 1.9 pg mL–1 and 2.5 ± 0.8 pg mL–1, respectively), this was not significantly different. Ovaries treated and cultured on a membrane without VEGFAxxxb produced more E2 (22.5 ± 12.2 pg mL–1) than ovaries treated on a membrane and cultured in an alginate bead, with (3.9 ± 1.9 pg mL–1) or without (1.8 ± 1.2 pg mL–1) VEGFAxxxb; this was not significantly different from ovaries treated and cultured on a membrane with VEGFAxxxb (4.6 ± 4.6 pg mL–1). In summary, primordial follicles can be induced to activate and grow in vitro using a PTEN inhibitor, as evidenced by the upregulated expression of follicle growth genes; VEGFAxxxb also increases expression of these genes. Treatment in a bead compared with a membrane appears to be advantageous for gene expression. However, culture on a membrane best supported oestradiol production, regardless of the presence of VEGFxxxb. These are critical pieces in the development of a successful culture system in which primordial follicles can be activated and functional growth supported in vitro.


Reproduction ◽  
2007 ◽  
Vol 133 (1) ◽  
pp. 107-116 ◽  
Author(s):  
Chuck R Greenfeld ◽  
Janice K Babus ◽  
Priscilla A Furth ◽  
Sam Marion ◽  
Patricia B Hoyer ◽  
...  

Mammalian females are endowed with a finite number of primordial follicles at birth or shortly thereafter. Immediately following the formation of the primordial follicle pool, cohorts of these follicles are recruited to begin growth, and this recruitment continues until the primordial follicle population is depleted. Once recruited, a follicle will either grow and ovulate or undergo atresia. Follicle atresia results from the apoptotic death of follicular cells. Members of the BCL-2 family of proteins are important regulators of apoptosis in most cells including in the ovary. Here, we tested the hypothesis that the proapoptotic BAX is an important regulator of follicle survival. We used a variety of histological and biochemical techniques to investigate the impact ofBaxdeletion on follicle growth and death. We observed that theBaxdeletion results in delayed vaginal opening and altered follicular growth. Young adultBax-deficient ovaries contained increased numbers of primordial follicles and a trend towards reduced numbers of growing follicles.Baxdeficiency led to a reduction in average litter size, and also a reduction in the number of oocytes ovulated in response to exogenous gonadotropins. In contrast,Baxdeficiency did not alter follicle atresia. In conclusion, BAX appears to be an important regulator of follicle growth, but is dispensable for follicle atresia in mice.


2021 ◽  
Vol 22 (12) ◽  
pp. 6570
Author(s):  
Yue Lv ◽  
Rui-Can Cao ◽  
Hong-Bin Liu ◽  
Xian-Wei Su ◽  
Gang Lu ◽  
...  

A better understanding of the mechanism of primordial follicle activation will help us better understand the causes of premature ovarian insufficiency (POI), and will help us identify new drugs that can be applied to the clinical treatment of infertility. In this study, single oocytes were isolated from primordial and primary follicles, and were used for gene profiling with TaqMan array cards. Bioinformatics analysis was performed on the gene expression data, and Ingenuity Pathway Analysis was used to analyze and predict drugs that affect follicle activation. An ovarian in vitro culture system was used to verify the function of the drug candidates, and we found that curcumin maintains the ovarian reserve. Long-term treatment with 100 mg/kg curcumin improved the ovarian reserve indicators of AMH, FSH, and estradiol in aging mice. Mechanistic studies show that curcumin can affect the translocation of FOXO3, thereby inhibiting the PTEN-AKT-FOXO3a pathway and protecting primordial follicles from overactivation. These results suggest that curcumin is a potential drug for the treatment of POI patients and for fertility preservation.


Reproduction ◽  
2021 ◽  
Author(s):  
Kouji Komatsu ◽  
Wei Wei ◽  
Tomohiko Murase ◽  
Satoru Masubuchi

This study aimed to clarify the physiological mechanism regulating the growth of primordial follicles in mouse ovaries. In a previous study, we found that increasing the fetal bovine serum concentration in the culture medium promoted the growth of primordial follicles in cultured postnatal day 0 ovaries but not in cultured postnatal day 4 ovaries. Based on these results, we hypothesized that the regulatory system repressing the growth of primordial follicles is established in postnatal day 4 ovaries. To confirm this hypothesis, microarray analysis of postnatal day 0 and 4 ovaries was performed. The results revealed that the expression of mRNA of stefin A homologs increased in postnatal day 4 ovaries. Stefin A belonging to the type 1 cystatin superfamily is an inhibitor of cysteine cathepsins. Consistently, the inhibitor of cathepsins repressed the growth of primordial follicles in cultured postnatal day 0 ovaries. Furthermore, we found that 17β-estradiol promoted the expression of mRNA of stefin A homologs in cultured ovaries and repressed the growth of primordial follicles. Our results demonstrate that 17β-estradiol and cathepsins regulate the growth of primordial follicles in mouse ovaries.


2020 ◽  
Vol 2020 (4) ◽  
Author(s):  
C De Roo ◽  
S Lierman ◽  
K Tilleman ◽  
P De Sutter

Abstract STUDY QUESTION What is the role of the Hippo and PI3K/Akt pathway in follicles during ovarian tissue culture in tissue derived from oncological patients and transgender men? SUMMARY ANSWER Results highlight a Hippo pathway driven primordial follicle activation in vitro, predominantly from Day 0 to Day 4. WHAT IS KNOWN ALREADY In-vitro ovarian tissue culture aims at activating and maturing primordial follicles for fertility restoration in patients with a threatened ovarian reserve. Not all patients are eligible for ovarian cortex transplantation and therefore several groups are attempting to culture ovarian tissue in-vitro. Cortex fragmentation disrupts the Hippo pathway, leading to increased expression of downstream growth factors and follicle growth. The PI3K/Akt pathway is considered the intracellular pathway to where different extracellular factors involved in primordial follicle activation in-vivo converge. In order to optimise current ovarian tissue culture models, information on progression of these pathways during tissue culture is mandatory. STUDY DESIGN, SIZE, DURATION The first step of a multistep cortex culture system was performed using 144 ovarian cortex pieces from a total of six patients. Per patient, 24 cortical strips were cultured for 6 days and six pieces per patient were collected for downstream analysis of follicle development and Hippo and PI3K/Akt pathway targets every second day. PARTICIPANTS/MATERIALS, SETTING, METHODS Ovarian tissue was obtained from oncological (N = 3; 28.67 ± 4.51 years) and transgender (N = 3; 23.33 ± 1.53 years) patients. Follicles were analysed using haematoxylin-eosin staining and pathways were studied using immunohistochemistry and precise follicle excision by laser capture micro-dissection for RT-qPCR analysis. MIQE guidelines for RT-qPCR were pursued. Reference gene selection (GAPDH, RPL3A, 18s rRNA) was performed using GeNorm Reference Gene Selection Kit. Statistical analysis was conducted with IBM SPSS Statistics 23 (Poisson regression, negative binomial regression, ANOVA and paired t-test). MAIN RESULTS AND THE ROLE OF CHANCE Immunohistochemical analysis confirmed a Hippo pathway driven primordial follicle activation due to mechanical manipulation of the cortical strips. Ovarian tissue preparation and culture induced the inhibitory phosphorylated Yes-associated protein (pYAP) to disappear in granulosa cells of primordial follicles on Day 2. The stimulatory YAP on the contrary appeared in primordial granulosa cells over increasing culture days. Looking at the YAP target connective tissue growth factor (CTGF), a significantly up-regulated CTGF was noted in primordial follicles when comparing Day 2 and Day 4 (ratio Day 2/4 = 0.082; P &lt; 0.05), clearly showing an effect on the Hippo pathway in primordial follicles during tissue culture. Follicle classification showed a significant drop in estimated primordial follicle counts in the oncological cohort (−78%; P = 0.021) on Day 2 and in the transgender cohort on Day 4 (−634%; P = 0.008). Intermediate follicle counts showed a non-significant increasing trend to during culture and this follicle recruitment and growth resulted in a significant rise in estimated primary follicle counts on Day 6 in oncological patients (170%; P = 0.025) and, although limited in absolute numbers, a significant increase in secondary follicles on Day 4 (367%; P = 0.021) in the transgender cohort. Subsequent antral follicle development could not be observed. LIMITATIONS, REASONS FOR CAUTION A limitation is the small sample size, inherent to this study subject, especially as a large amount of tissue was needed per patient to reduce inter-patient variation in different downstream analysis techniques. A particular and specific weakness of this study is the inability to include an age-matched control group. WIDER IMPLICATIONS OF THE FINDINGS These findings support an adapted tissue preparation for Hippo pathway disruption and a shorter first phase of tissue culture. This work may also have implications for transplantation of cryopreserved tissue as larger strips (and thus slower burnout due to less Hippo pathway disruption) could be a benefit. STUDY FUNDING/COMPETING INTEREST(S) This research was financially supported by the Foundation Against Cancer (Stichting tegen Kanker, TBMT001816N), the Flemish Foundation of Scientific Research (FWO Vlaanderen, FWO G0.065.11N10) and the Gender Identity Research and Education Society (GIRES) foundation. The authors declare no competing interests. TRIAL REGISTRATION NUMBER N/A.


2020 ◽  
Author(s):  
V. Praveen Chakravarthi ◽  
Subhra Ghosh ◽  
Katherine F. Roby ◽  
Michael W. Wolfe ◽  
M. A. Karim Rumi

AbstractOver the entire reproductive lifespan in mammals, a fixed number of primordial follicles serve as the source of mature oocytes. Uncontrolled and excessive activation of primordial follicles can lead to depletion of the ovarian reserve. We observed that disruption of ESR2-signaling results in increased activation of primordial follicles in Esr2-null (Esr2-/-) rats. However, follicle assembly was unaffected, and the total number of follicles remained comparable between neonatal wildtype and Esr2-/- ovaries. While the activated follicle counts were increased in Esr2-/- ovary, the number of primordial follicles were markedly decreased. Excessive recruitment of primordial follicles led to premature ovarian senescence in Esr2-/- rats and was associated with reduced levels of serum AMH and estradiol. Disruption of ESR2-signaling through administration of a selective antagonist (PHTPP) increased the number of activated follicles in wildtype rats, whereas a selective agonist (DPN) decreased follicle activation. In contrast, primordial follicle activation was not increased in the absence of ESR1 indicating that the regulation of primordial follicle activation is ESR2-specific. Follicle activation was also increased in Esr2-mutants lacking the DNA-binding domain, suggesting a role for the canonical transcriptional activation function. Both primordial and activated follicles express ESR2 suggesting a direct regulatory role for ESR2 within these follicles. We also detected that loss of ESR2 augmented the activation of AKT, ERK and mTOR pathways. Our results indicate that the lack of ESR2 upregulated both granulosa and oocyte factors, which can facilitate AKT and mTOR activation in Esr2-/- ovaries leading to increased activation of primordial follicles.


2020 ◽  
Vol 20 (3) ◽  
pp. 899-917
Author(s):  
Monika Hułas-Stasiak ◽  
Joanna Jakubowicz-Gil ◽  
Piotr Dobrowolski ◽  
Małgorzata Grzesiak ◽  
Siemowit Muszyński ◽  
...  

AbstractΒ-hydroxy-β-methylbutyrate (HMB) is one of the leucine metabolites with protein anabolic effects which makes it very popular among athletes. Previously, it was shown that HMB administered during the prenatal period reduced the pool of primordial follicles and increased the proportion of developing follicles in newborn piglets. This work is a further step to understand these morphological alterations. Therefore, the aim of this study was to examine the effect of prenatal HMB treatment on the expression of the Kit ligand, BMP-4, bFGF, and the IGF-1/IGF-1R system which are the main growth factors controlling follicular development. Excised ovaries from 12 newborn piglets, originated from the control (n=6) and HMB-treated (n=6) sows were used for immunohistochemical and western-blot analysis. The tested proteins were localized within egg nests and ovarian follicles. Furthermore, the western-blot assay indicated higher BMP-4, Kit ligand, and IGF-1R expression, while the level of bFGF and IGF-1 proteins decreased after HMB dietary treatment. These findings show that HMB included into sow diet can modulate the expression of growth factors and thereby alter ovarian morphology in offspring. Therefore, this study opens a discussion about the benefits and risks of the diet supplemented with HMB and its potential application in medicine and animal husbandry, and further research is necessary in this area.


Sign in / Sign up

Export Citation Format

Share Document