Next Article in Journal
Plantar Mechanical Stimulation Maintains Slow Myosin Expression in Disused Rat Soleus Muscle via NO-Dependent Signaling
Next Article in Special Issue
The Amniotic Fluid Cell-Free Transcriptome Provides Novel Information about Fetal Development and Placental Cellular Dynamics
Previous Article in Journal
A Novel Multiprotein Bridging Factor 1-Like Protein Induces Cyst Wall Protein Gene Expression and Cyst Differentiation in Giardia lamblia
Previous Article in Special Issue
FTIR Spectroscopy to Reveal Lipid and Protein Changes Induced on Sperm by Capacitation: Bases for an Improvement of Sample Selection in ART
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ovarian Aging: Molecular Mechanisms and Medical Management

by
Jan Tesarik
*,
Maribel Galán-Lázaro
and
Raquel Mendoza-Tesarik
MARGen Clinic, 18006 Granada, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(3), 1371; https://doi.org/10.3390/ijms22031371
Submission received: 21 December 2020 / Revised: 26 January 2021 / Accepted: 26 January 2021 / Published: 29 January 2021
(This article belongs to the Special Issue Advanced Techniques in Reproductive Medicine Research)

Abstract

:
This is a short review of the basic molecular mechanisms of ovarian aging, written with a particular focus on the use of this data to improve the diagnostic and therapeutic protocols both for women affected by physiological (age-related) ovarian decay and for those suffering premature ovarian insufficiency. Ovarian aging has a genetic basis that conditions the ovarian activity via a plethora of cell-signaling pathways that control the functions of different types of cells in the ovary. There are various factors that can influence these pathways so as to reduce their efficiency. Oxidative stress, often related to mitochondrial dysfunction, leading to the apoptosis of ovarian cells, can be at the origin of vicious circles in which the primary cause feeds back other abnormalities, resulting in an overall decline in the ovarian activity and in the quantity and quality of oocytes. The correct diagnosis of the molecular mechanisms involved in ovarian aging can serve to design treatment strategies that can slow down ovarian decay and increase the quantity and quality of oocytes that can be obtained for an in vitro fertilization attempt. The available treatment options include the use of antioxidants, melatonin, growth hormones, and mitochondrial therapies. All of these treatments have to be considered in the context of each couple’s history and current clinical condition, and a customized (patient-tailored) treatment protocol is to be elaborated.

Graphical Abstract

1. Introduction

The importance of issues related to ovarian aging has been increasing progressively over the past several decades since, increasingly, more couples in all developed countries choose to postpone parenthood to more advanced female ages [1]. This trend is associated with an increasing rate of aneuploidy in oocytes, causing chromosomal abnormalities in embryos resulting from natural conception, conventional in vitro fertilization (IVF), or intracytoplasmic sperm injection (ICSI) [2,3]. On the other hand, no association has been found between advanced male age and aneuploidy rates in embryos derived from IVF/ICSI attempts using oocytes from young donors [1].
The mechanisms involved in ovarian aging are not completely understood and appear to be multifactorial. A better knowledge of the factors and mechanisms causing age-related or premature ovarian decay is needed to optimize diagnostic tests and tune treatment options so as to reflect the individual condition of each couple. This minireview resumes different factors causing ovarian decay and their respective molecular mechanisms of action with the aim of elaborating a patient-tailored treatment regimen for each individual couple.

2. Methods

Works used for this minireview were found by searching MEDLINE (PubMed) from 1995 to 2020. A combination of medical subject headings and keywords was used to generate a subset of citations as following (in alphabetical order): (i) antioxidants, (ii) apoptosis, (iii) epigenetic, (iv) genetic, (v) mitochondria, (vi) ovarian aging, (vii) oxidative stress.

3. Molecular Mechanisms

The age of menopause is an inheritable trait, and the age at which primary ovarian failure occurs has a strong genetic component [4,5]. However, not all ovarian failures are primary, and different associated pathologies can play a role. Some of them appear to be related to defective DNA repair pathways [6].

3.1. Genetic Basis

Genetic factors can influence ovarian function selectively (primary ovarian insufficiency) or as part of the symptomatology of disorders implicated in other pathologies.

3.1.1. Primary Ovarian Insufficiency

Primary ovarian insufficiency (POI) is the most frequent cause of early menopause, which occurs in about 10% of women before 45 years of age and in 1–2% before 40 years [4], while fertility impairment starts around 20 years before the menopause [7]. Chromosome X structural abnormalities and X-autosome translocations can be at the origin of some cases of POI [6]. As for single-gene perturbations, several genes have been suggested to be implicated in POI (Table 1), some of them located on the X-chromosome and others on autosomes [4,8,9].
The former group includes bone morphogenetic protein 15 (BMP15) (Xp11.2), progesterone receptor membrane component 1 (PGRMC1) (Xq22-q24), androgen receptor (AR) (Xq12), forkhead box O4 (FOXO4) (Xq13.1), premature ovarian failure 1B (POF1B) (Xq21.2), dachshund family transcription factor 2 (DACH2) (Xq21.3), and fragile X mental retardation 1 (FMR1) (Xq27.3).
The genes on autosomes include growth differentiation factor 9 (GDF9) (5q31.1); folliculogenesis-specific bHLH transcription factor (FIGLA) (2p13.3); newborn ovary homeobox gene (NOBOX) (7q35); nuclear receptor subfamily 5, group A, member 1 (NR5A1); steroidogenic factor-1 (SF-1) (9q33); FSH receptor (FSHR) (2p21-p16); TGF beta receptor III (TGFBR3) (1p33-p32); G protein-coupled receptor 3 (GPR3) (1p36.1-p35); wingless-type MMTV integration site family member 4 (WNT4) (1p36.23-p35.1); inhibins: inhibin alpha (INHA) (2q35), inhibin beta A (INHBA) (7p15-p13), inhibin beta B (INHBB) (2cen-q13); POU class 5 homeobox 1 (POU5F1) (6p21.31); MutS homolog 4 (MSH4) (1p31) and MSH5 (6p21.3); forkhead box O3 (FOXO3) (6q21); cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-terminar domain 2 (CITED2) (6q23.3); spermatogenesis-and oogenesis-specific basic helix-loop-helix transcription factor 1 (SOHLH1) ((9q34.3) and SOHLH2 (13q13.3); phosphatase and tensin homolog (PTEN) (10q23.3); Drosophila nanos homologs 1, 2, and 3: NANOS 1 (10q26.11), NANOS2 (19q13.32), NANOS 3 19p13.13); cyclin-dependent kinase inhibitor 1B (CDKN1B) (12p13.1-p12); anti-Mullerian hormone receptor type II (AMHR2) (12q13); forkhead box O1 (FOXO1) (13q14.1); spalt-like transcription factor 4 (SALL4) (20q13.2); and DNA meiotic recombinase 1 (DMC1) (22q13.1).
However, the implication of some of these genes is suspected rather than confirmed. Those of them with the strongest evidence for playing a role in POI are shown in Table 1.
Most of these genes (Table 1) are known to be somehow related to oogenesis during its different phases, from the fetal period throughout postnatal life until the final phases of meiotic maturation, and to regulate essential events in human oogenesis [4,5]. These include female sex determination and differentiation (WNT4), the formation of primordial follicles and the coordinated expression of zona pellucida genes (FIGLA), the transition from primordial to growing follicles (NOBOX, FOXO3, PTEN), the hormone-dependent phase of follicular growth (FSHR), the maintenance of meiotic arrest in antral follicles until the luteinizing hormone (LH) surge (GPR3), DNA mismatch repair during meiotic recombination (MSH4, MSH5), the apoptosis of ovarian cells (PGRMC1), granulosa cell function (FOXO1), and the repair of DNA damage arising from defective meiotic divisions (DMC1) [4,6,8,9] (Table 1). However, the incidence of anomalies in some of these genes among women suffering from POI is extremely low, and perturbations of other genes are mostly related to specific ethnic groups [10]. Thus, only a few of these genes, such as FMR1 premutation, BMP15, GDF9, and FSHR, have been incorporated as diagnostic biomarkers [5,11,12]. More research is needed to use more genes as routine diagnostic tools.

3.1.2. Ovarian Insufficiency due to Mendelian Disorders Implicated in Other Pathologies

Distinct from non-syndromic POI, pleiotropic Mendelian disorders, including fragile X syndrome: familial mental retardation 1 (FMR1); (Xq27.3), blepharophimosis-ptosis-epicanthus syndrome (BPES): forkhead box L2 (FOXL2) (3q23); galactosemia: galactose-1-phosphate uridyl transferase (GALT) (9p13); carbohydrate-deficient glycoprotein syndrome type 1: phosphomannomutase 2 (PMM2) (16p13), may manifest POI as part of their phenotypic spectrum [4]. There are some other pleiotropic Mendelian disorders suspected to cause POI, but a definitive confirmation of this association is still lacking.

3.1.3. Gene Mutations Affecting Mitochondrial Function

Genes governing mitochondrial functions may be located in the nucleus or in the mitochondria themselves. Many different nuclear genes affecting mitochondrial function are known nowadays, and according to current experience most gene mutations impairing mitochondrial function are nuclear ones, as reviewed in [4]. Both oocyte and ovarian cell mitochondria are important for the correct folliculogenesis and oocyte maturation [13,14,15,16]. As to the risk of mitochondrial DNA deletions, it was shown to be affected by the global secondary structure of the mitochondrial genome [17].

3.2. Cell-Signaling Pathways

The main cell-signaling pathways involved in physiological (age-related) ovarian failure and POI are those involved in cell protection against oxidative stress. A recent study using single-cell transcriptomic analysis of ovaries from young and aged non-human primates identified seven ovarian cell types with distinct gene-expression signatures, including oocyte and six different somatic cells, and identified the disturbance of antioxidant signaling specific to early-stage oocytes and granulosa cells [9]. The further analysis of cell-type-specific aging-associated transcriptional changes uncovered age-related disturbances of antioxidant signaling specific to early-stage oocytes and granulosa cells [9]. The authors have completed their observations on non-human primates with those on human granulosa cells obtained from follicular fluid samples aspirated from patients undergoing an IVF attempt. Consistent with the results in monkeys, human granulosa cells exhibited the age-related downregulation of the transcription of three genes involved in antioxidative pathways—IDH1, PRDX4, and NDUFB10—and this phenomenon was accompanied by an increase in reactive oxygen species and apoptosis in the granulosa cells [18], indicative of oxidative damage as a crucial factor in ovarian functional decay.
Similar to the primates, an age-related increase in oxidative damage and a decrease in antioxidant gene expression was previously observed in mice [14]. The oxidative damage of mouse granulosa cells was reported to impair the supply of ATP and the mitochondrial gene expression, which are required not only for the proliferation but also the differentiation of granulosa cells during follicular development [13]. Moreover, aging-related mitochondrial (mt) DNA instability also leads to an accumulation of mtDNA mutations in the oocyte, leading to the deterioration of oocyte quality in terms of competence and the risk of transmitting mitochondrial abnormalities to offspring [14]. As mentioned above, the risk of mtDNA damage is also conditioned by epigenetic factors, especially the global secondary structure of the mitochondrial genome; certain patterns of the global secondary structure of the human single-stranded heavy chain of mtDNA make the DNA molecule more prone to deletions than others [17].
Mitochondrial damage leads to the activation of apoptotic pathways in granulosa cells, which, in turn, decreases the expression of aromatase, which is required for the transformation of androgens to estrogens, thus leading to the prevalence of androgens over estrogens within the follicles [18]. Androgens and estrogens present in follicular fluid exert rapid non-genomic effects on maturing human oocytes, affecting oocyte cytoplasmic maturation and postfertilization developmental potential rather than the completion of meiosis [19]. This can explain why age-related or premature disturbances of antioxidative pathways reduce oocyte quality even in cases in which the number of mature oocytes recovered for IVF is not affected.
New data suggest that POI may be of polygenic origin, and that overlap exists between the genetic backgrounds of diminished ovarian reserves and POI [20]. Whole-exome sequencing and bioinformatics analysis may become a useful clinical tool for etiological diagnosis and risk prediction for affected women in the future [20]. In addition, array comparative genomic hybridization or specific next-generation sequencing panels should be considered to identify chromosomal deletions/duplications under karyotype resolution or other pathogenic variants in specific genes associated with POI. This is particularly important in patients with first-or second-degree relatives also affected with POI, improving their reproductive and genetic counseling [21].
Moreover, even with the same genetic background, with the use of multiple systems biologic approaches to compare developmental stages in the early human embryo with single-cell transcript data from blastomeres, it was shown that blastomeres considered to be totipotent were not transcriptionally equivalent [22].

4. Clinical Management

4.1. Diagnosis

A woman’s age is the basic predictor of the degree of ovarian aging. However, ovarian aging can develop prematurely, and specific diagnostic methods are needed to detect this condition. There are two types of ovarian aging manifestations with respect to the ovarian ability to produce oocytes: a quantitative one and a qualitative one. To detect the former, a combination of antral follicle count (AFC), determined by vaginal ultrasound scan at the beginning of the menstrual cycle, and the determination of serum anti-Mullerian hormone (AMH) concentration, which can be performed at any time during the menstrual cycle [23], is used.
By contrast, oocyte quality is not necessarily related to AFC and serum AMH. Premature decay of oocyte quality is mainly related to a low-for-age production of growth hormone (GH) [24]. Due to the pulsatile pattern of GH secretion, insulin-like growth factor-1 (IGF-1), which does reflect the GH secretion pattern, but with a less pronounced pulsatility, was suggested to identify young patients with a premature decay of oocyte quality who could benefit from treatment with GH during ovarian stimulation [25]. The data presented show that the ‘GH/IGF-1’ age can be more than 20 years higher than the chronological age in some young women, and it was this group of patients who appeared to be more likely to benefit from GH administration during ovarian stimulation, although the authors suggested that larger prospective studies were needed to confirm this assumption [25].
There are only a few clues to detect premature ovarian aging in addition to the above criteria, and further research is warranted to detect more molecular markers that could guide the clinician to propose the best treatment regimen.

4.2. Treatment

In view of the fact that oxidative stress is the main factor involved in ovarian aging, it can be assumed that agents reducing oxidative damage represent the first-line choice. These agents can be direct antioxidants or molecules affecting cell-signaling pathways involved in the antioxidant defense of ovarian cells (Table 2). Some molecules combine both of the above activities.

4.2.1. GH

GH administration during ovarian stimulation was the first treatment shown to be beneficial in older women. A randomized controlled trial, conducted in a group of 100 women of >40 years undergoing assisted reproduction treatment and randomized between a GH treatment group and a placebo group, showed significantly higher delivery and live birth rates in the GH arm as compared to the placebo arm [26]. Subsequent studies confirmed these findings and extended the use of GH treatment also to younger women with POI [27,28,29,30,31]. This effect of GH is at least partly due to the alleviation of oxidative stress in the ovary, an effect previously described in some other organs [25]. Though it is not a direct antioxidant, GH intervenes in the cell-signaling pathways involved in cellular defense against oxidative stress [28], and adult GH deficiency causes an inadequate reactivity of cells against radical production [29]. This can explain why age-related or premature GH deficiency contributes to ovarian decay, even in cases where it is primarily due to other causes. Hence, GH can be used as an adjuvant treatment during ovarian stimulation in women with both age-related ovarian decay and POI. This conclusion was drawn from the results of 13 articles of a special research topic on the role of GH in reproduction, edited by Jan Tesarik, John Yovich, and Yves Menezo, and published in Frontiers in Endocrinology, reviewed in Tesarik et al. in 2021 [32].

4.2.2. Melatonin

Melatonin is an example par excellence of a molecule acting as a direct antioxidant and a modulator of systems protecting cells against oxidative stress. First introduced into reproductive medicine to treat infertility caused by endometriosis and adenomyosis [33,34], it has proven its usefulness in many more indications, including the protection against the contraction of COVID-19 [35]. Since then, new data have emerged showing the possibilities of melatonin in preventing ovarian aging [36]. Even though the mechanism underlying the anti-aging effect of melatonin in the human ovary still needs to be fully explained, the administration of melatonin, lacking serious side-effects and providing additional benefits to patients treated with it [36], is clearly indicated in women with age-related ovarian decay or POI.

4.2.3. Other Antioxidants

Since ovarian aging is mainly due to oxidative stress, superimposed on the existing genetic makeup, any antioxidant agent can be supposed to improve IVF results in older women and in young women suffering from POI [37,38,39]. Unfortunately, there are only a few clinical studies that can conclusively support this reasoning. Coenzyme Q10 (CoQ10) is the antioxidant that accumulates the most evidence in favor of its use in the treatment of women with diminished ovarian reserves [37,38], a conclusion corroborated by a recent randomized control trial [39]. However, though potentially less efficient, any antioxidant agent, such as vitamins C and E and folic acid, can be of help [40].

4.2.4. Mitochondrial Therapy

The issue of mitochondrial health has been widely debated with respect to the oocyte [41]. Nevertheless, data obtained from a study in an animal model (mouse) indicate that the granulosa cell mitochondria are no less important for the correct oocyte maturation than those of the oocyte itself [15]. Antioxidant agents, such as melatonin, coenzyme Q10, or vitamins C and E (see above) can improve this condition. This can explain why antioxidant pretreatment (coenzyme Q10) improves the ovarian response to gonadotropin stimulation and embryo quality in low-prognosis young women [37,38,39].
On the other hand, the accumulation of mitochondrial DNA mutations/deletions in the oocyte, in addition to jeopardizing oocyte developmental potential, can also compromise the health of the offspring [42]. Optimal mitochondrial function is required for oocyte maturation, fertilization, and embryonic development. Improvement of the mitochondrial function, either through the use of small molecules or procedures involving mitochondrial transfers, could lead to better fertility outcomes. Moreover, mitochondrial replacement procedures could open a new page in the treatment of mitochondrial diseases.
Mitochondrial transfer from a healthy donor oocyte to the patient’s oocyte is a possible solution, practiced from the late 1990s [42,43,44,45] but thereafter banned in most countries. This technique can be performed in two different ways: first, by injection of a small amount of donor oocyte cytoplasm into the patient’s oocytes [42,43,44]; second, by transferring the metaphase chromosomes, associated with the meiotic spindle, from the patient’s oocytes to previously enucleated donor oocytes [45]. Curiously, even though the latter technique results in a much higher proportion of “healthy” mitochondria in the reconstructed oocytes as compared to the former one, the efficiency of both techniques appeared similar; more than 40% of live births in young women with previous implantation failures, and several tens of births were obtained with both of them [46,47]. It has to be stressed, however, that the original indication of both of these techniques was focused on the recurrent failure of embryo development and implantation in young women, rather than on the avoidance of mitochondrial disease transmission. In addition to mitochondria, ooplasm also contains other developmentally important molecules that can be deficient in patients’ oocytes. One of them is stored maternal mRNA which is crucial for guiding human embryo development until the 4-to 8-cell stage, when the first signs of embryonic genome expression can be detected [48,49,50,51]. However, stored maternal mRNA is also involved in the control of relatively late stages of human preimplantation development, after the activation of embryonic gene expression, when the two mRNA sources act together to regulate the differentiation of the first two embryonic tissues, the inner cell mass, and the trophectoderm [51].
While still banned in the United States, the technique of mitochondrial replacement therapy, using a spindle-chromosome complex transfer from the patient’s oocytes to enucleated donor oocytes, was used with success by a team in a US clinic in Mexico, where there is no legal restriction with regard to this technique, to avoid the mother-to-offspring transmission of a heritable mitochondrial disease, the Leigh syndrome [52]. The authors chose the previously described technique of nuclear transfer, with slight modifications [53], rather than ooplasmic injection. This technique is currently used, in countries in which it is legally possible, for its initial indication: the repeated failure of embryonic development in young couples with male partners having normal spermatozoa, without strict limitations to previously detected mitochondrial DNA abnormalities.

4.2.5. Patient-Tailored (Customized) Treatment Protocols

In order to be able to decide between different treatment options, the choice of the therapy to be used must be made on the basis of a complete diagnosis of both the male and female partner of each couple. The treatment choice should not be based merely on the primary cause of infertility, but should also take into account all possible secondary contributing factors. Basic guidelines for this approach were published under the name “CARE” (Customized Assisted Reproduction Enhancement) [54]. In bad-prognosis patients, this individualized approach seems to be more efficient as compared with standard protocols [55]. Further prospective studies are needed to definitively confirm this thesis.

5. Conclusions

All the available data lead to the conclusion that ovarian aging is mainly due to oxidative stress. While the external factors leading to oxidative stress might be similar, the damage produced is patient-dependent due to genetically programmed defense mechanisms. Deficiencies of these mechanisms can be caused by mutations/deletions of both nuclear and mitochondrial genes. Independent of the genetic background (which is impossible to resolve at present), the clinical manifestations can be alleviated with treatments using direct antioxidants (e.g., vitamins C and E, coenzyme Q10), agents affecting the cell response against oxidative stress (e.g., growth hormone), or those combining both of these activities (e.g., melatonin). If mitochondrial DNA mutations occur in the oocyte, mitochondrial replacement therapy can resolve the problem. Independent of the results of the individual diagnostic tests performed, a synthetic view is needed to propose a customized therapeutic plan for each infertile couple.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dviri, M.; Madjunkova, S.; Koziarz, A.; Antes, R.; Abramov, R.; Mashiach, J.; Moskovtsev, S.; Kuznyetsova, I.; Librach, C. Is there a correlation between paternal age and aneuploidy rate? An analysis of 3,118 embryos derived from young egg donors. Fertil. Steril. 2020, 114, 293–300. [Google Scholar] [CrossRef] [PubMed]
  2. Franasiak, J.M.; Forman, E.J.; Hong, K.H.; Werner, M.D.; Upham, K.M.; Treff, N.R.; Scott, R.T., Jr. The nature of aneuploidy with increasing age of the female partner: A review of 15,169 consecutive trophectoderm biopsies evaluated with comprehensive chromosomal screening. Fertil. Steril. 2014, 101, 656–663.e1. [Google Scholar] [CrossRef] [PubMed]
  3. Demko, Z.P.; Simon, A.L.; McCoy, R.C.; Petrov, D.A.; Rabinowitz, M. Effects of maternal age on euploidy rates in a large cohort of embryos analyzed with 24-chromosome single-nucleotide polymorphism-based preimplantation genetic screening. Fertil. Steril. 2016, 105, 1307–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Rossetti, R.; Ferrari, I.; Bonomi, M.; Persani, L. Genetics of primary ovarian insufficiency. Clin. Genet. 2017, 91, 183–198. [Google Scholar] [CrossRef] [Green Version]
  5. Ma, L.; Lu, H.; Chen, R.; Wu, M.; Jin, Y.; Zhang, J.; Wang, S. Identification of key genes and potential new biomarkers for ovarian aging: A study based on RNA-sequencing data. Front. Genet. 2020, 11, 590660. [Google Scholar] [CrossRef]
  6. Turan, V.; Oktay, K. BRCA-related ATM-mediated DNA double-strand break repair and ovarian aging. Hum. Reprod. Update 2020, 26, 43–57. [Google Scholar] [CrossRef]
  7. te Velde, E.R.; Pearson, P.L. The variability of female reproductive ageing. Hum. Reprod. Update 2002, 8, 141–154. [Google Scholar] [CrossRef]
  8. Qin, Y.; Jiao, X.; Simpson, J.L.; Chen, Z.J. Genetics of primary ovarian insufficiency: New developments and opportunities. Hum. Reprod. Update 2015, 21, 787–808. [Google Scholar] [CrossRef]
  9. Wang, S.; Zheng, Y.; Li, J.; Yu, Y.; Zhang, W.; Song, M.; Liu, Z.; Min, Z.; Hu, H.; Jing, Y.; et al. Single-cell transcriptomic atlas of primate ovarian aging. Cell 2020, 180, 585–600.e19. [Google Scholar] [CrossRef]
  10. Jin, M.; Yu, Y.; Huang, H. An update on primary ovarian insufficiency. Sci. China Life Sci. 2012, 55, 677–686. [Google Scholar] [CrossRef] [Green Version]
  11. Fortuño, C.; Labarta, E. Genetics of primary ovarian insufficiency: A review. J. Assist. Reprod. Genet. 2014, 31, 1573–1585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Luoma, P.; Melberg, A.; Rinne, J.O.; Kaukonen, J.A.; Nupponen, N.N.; Chalmers, R.M.; Oldfors, A.; Rautakorpi, I.; Peltonen, L.; Majamaa, K.; et al. Parkinsonism, premature menopause, and mitochondrial DNA polymerase gamma mutations: Clinical and molecular genetic study. Lancet 2004, 364, 875–882. [Google Scholar] [CrossRef]
  13. Pierce, S.B.; Gersak, K.; Michaelson-Cohen, R.; Walsh, T.; Lee, M.K.; Malach, D.; Klevit, R.E.; King, M.C.; Levy-Lahad, E. Mutations in LARS2, encoding mitochondrial leucyl-tRNA synthetase, lead to premature ovarian failure and hearing loss in Perrault syndrome. Am. J. Hum. Genet. 2013, 92, 614–620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lim, J.; Luderer, U. Oxidative damage increases and antioxidant gene expression decreases with aging in the mouse ovary. Biol. Reprod. 2011, 84, 775–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hoque, S.A.M.; Kawai, T.; Zhu, Z.; Shimada, M. Mitochondrial protein turnover is critical for granulosa cell proliferation and differentiation in antral follicles. J. Endocr. Soc. 2019, 3, 324–339. [Google Scholar] [CrossRef] [Green Version]
  16. May-Panloup, P.; Boucret, L.; Chao de la Barca, J.-M.; Desquiret-Dumas, V.; Ferré-L’Hotellier, V.; Morinière, C.; Descamps, P.; Procaccio, V.; Reynier, P. Ovarian ageing: The role of mitochondria in oocytes and follicles. Hum. Reprod. Update 2016, 22, 725–743. [Google Scholar] [CrossRef] [Green Version]
  17. Mikhailova, A.A.; Shamanskyi, V.; Ushakova, K.; Mikhailova, A.G.; Oreshkov, S.; Knorre, D.; Tretiakov, E.O.; Zazhytska, M.; Lukowski, S.W.; Liou, C.-W.; et al. Risk of mitochondrial deletions is affected by the global secondary structure of the mitochondrial genome. bioRxiv 2020, 603282. [Google Scholar] [CrossRef] [Green Version]
  18. Pizarro, B.M.; Cordeiro, A.; Reginatto, M.W.; Campos, S.P.C.; Mancebo, A.C.A.; Areas, P.C.F.; Antunes, R.A.; Souza, M.D.C.B.; Oliveira, K.J.; Bloise, F.F.; et al. Estradiol and progesterone levels are related to redox status in the follicular fluid during in vitro fertilization. J. Endocr. Soc. 2020, 4, bvaa064. [Google Scholar] [CrossRef]
  19. Tesarik, J.; Mendoza, C. Nongenomic effects of 17 beta-estradiol on maturing human oocytes: Relationship to oocyte developmental potential. J. Clin. Endocrinol. Metab. 1995, 80, 1438–1443. [Google Scholar] [CrossRef] [Green Version]
  20. Tang, R.; Yu, Q. Novel variants in women with premature ovarian function decline identified via whole-exome sequencing. J. Assist. Reprod. Genet. 2020, 37, 2487–2502. [Google Scholar] [CrossRef]
  21. Barros, F.; Carvalho, F.; Barros, A.; Dória, S. Premature ovarian insufficiency: Clinical orientations for genetic testing and genetic counseling. Porto Biomed. J. 2020, 5, e62. [Google Scholar] [CrossRef] [PubMed]
  22. Smith, H.L.; Stevens, A.; Minogue, B.; Sneddon, S.; Shaw, L.; Wood, L.; Adeniyi, T.; Xiao, H.; Lio, P.; Kimber, S.J.; et al. Systems based analysis of human embryos and gene networks involved in cell lineage allocation. BMC Genom. 2019, 20, 171. [Google Scholar] [CrossRef] [PubMed]
  23. Fleming, R.; Seifer, D.B.; Frattarelli, J.L.; Ruman, J. Assessing ovarian response: Antral follicle count versus anti-Müllerian hormone. Reprod. Biomed. Online 2015, 31, 486–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Yovich, J.L.; Regan, S.L.P.; Zaidi, S.; Keane, K.N. The concept of growth hormone deficiency affecting clinical prognosis in IVF. Front. Endocrinol. 2019, 10, 650. [Google Scholar] [CrossRef] [PubMed]
  25. Tesarik, J.; Mendoza-Tesarik, R. New criteria for the use of growth hormone in the treatment of female infertility: Minireview and a case series. EC Gynaecol. 2020, 9, 1–4. [Google Scholar]
  26. Tesarik, J.; Hazout, A.; Mendoza, C. Improvement of delivery and live birth rates after ICSI in women aged >40 years by ovarian co-stimulation with growth hormone. Hum. Reprod. 2005, 20, 2536–2541. [Google Scholar] [CrossRef] [Green Version]
  27. Li, X.L.; Wang, L.; Lv, F.; Huang, X.M.; Wang, L.P.; Pan, Y.; Zhang, X.M. The influence of different growth hormone addition protocols to poor ovarian responders on clinical outcomes in controlled ovary stimulation cycles: A systematic review and meta-analysis. Medicine 2017, 96, e6443. [Google Scholar] [CrossRef]
  28. Hart, R.J. Use of growth hormone in the IVF treatment of women with poor ovarian reserve. Front. Endocrinol. 2019, 10, 500. [Google Scholar] [CrossRef] [Green Version]
  29. Tesarik, J.; Galán-Lázaro, M.; Conde-López, C.; Chiara-Rapisarda, A.M.; Mendoza-Tesarik, R. The effect of GH administration on oocyte and zygote quality in young women with repeated implantation failure after IVF. Front. Endocrinol. 2020, 11, 519572. [Google Scholar] [CrossRef]
  30. Gong, Y.; Luo, S.; Fan, P.; Jin, S.; Zhu, H.; Deng, T.; Quan, Y.; Huang, W. Growth hormone alleviates oxidative stress and improves oocyte quality in Chinese women with polycystic ovary syndrome: A randomized controlled trial. Sci. Rep. 2020, 11, 18769. [Google Scholar] [CrossRef]
  31. Mancini, A.; Di Segni, C.; Bruno, C.; Olivieri, G.; Guidi, F.; Silvestrini, A.; Meucci, E.; Orlando, P.; Silvestri, S.; Tiano, L.; et al. Oxidative stress in adult growth hormone deficiency: Different plasma antioxidant patterns in comparison with metabolic syndrome. Endocrine 2018, 59, 130–136. [Google Scholar] [CrossRef] [PubMed]
  32. Tesarik, J.; Yovich, J.; Menezo, Y. Growth Hormone in Fertility and Infertility: Physiology, Pathology, Diagnosis and Treatment. Front. Endocrinol. 2020, in press. [Google Scholar]
  33. Tesarik, J.; Mendoza-Tesarik, R. Melatonin: The first noninvasive causal therapy for both endometriosis and adenomyosis? J. Gynecol. Women’s Health 2018, 12, 555829. [Google Scholar] [CrossRef]
  34. Mosher, A.A.; Tsoulis, M.W.; Lim, J.; Tan, C.; Agarwal, S.K.; Leyland, N.A.; Foster, W.G. Melatonin activity and receptor expression in endometrial tissue and endometriosis. Hum. Reprod. 2019, 34, 1215–1224. [Google Scholar] [CrossRef]
  35. Tesarik, J. Melatonin attenuates growth factor receptor signaling required for SARS-CoV-2 replication. Melatonin Res. 2020, 3, 534–537. [Google Scholar] [CrossRef]
  36. Tamura, H.; Jozaki, M.; Tanabe, M.; Shirafuta, Y.; Mihara, Y.; Shinagawa, M.; Tamura, I.; Maekawa, R.; Sato, S.; Taketani, T.; et al. Importance of melatonin in assisted reproductive technology and ovarian aging. Int. J. Mol. Sci. 2020, 21, 1135. [Google Scholar] [CrossRef] [Green Version]
  37. Budani, M.C.; Tiboni, G.M. Effects of Supplementation with Natural Antioxidants on Oocytes and Preimplantation Embryos. Antioxidants 2020, 9, 612. [Google Scholar] [CrossRef]
  38. Gat, I.; Blanco Mejia, S.; Balakier, H.; Librach, C.L.; Claessens, A.; Ryan, E.A. The use of coenzyme Q10 and DHEA during IUI and IVF cycles in patients with decreased ovarian reserve. Gynecol. Endocrinol. 2016, 32, 534–537. [Google Scholar] [CrossRef]
  39. Xu, Y.; Nisenblat, V.; Lu, C.; Li, R.; Qiao, J.; Zhen, X.; Wang, S. Pretreatment with coenzyme Q10 improves ovarian response and embryo quality in low-prognosis young women with decreased ovarian reserve: A randomized controlled trial. Reprod. Biol. Endocrinol. 2018, 16, 29. [Google Scholar] [CrossRef]
  40. Agarwal, A.; Durairajanayagam, D.; du Plessis, S.S. Utility of antioxidants during assisted reproductive techniques: An evidence based review. Reprod. Biol. Endocrinol. 2014, 12, 112. [Google Scholar] [CrossRef] [Green Version]
  41. Babayev, E.; Seli, E. Oocyte mitochondrial function and reproduction. Curr. Opin. Obstet. Gynecol. 2015, 27, 175–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Tachibana, M.; Kuno, T.; Yaegashi, N. Mitochondrial replacement therapy and assisted reproductive technology: A paradigm shift toward treatment of genetic diseases in gametes or in early embryos. Reprod. Med. Biol. 2018, 17, 421–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Cohen, J.; Scott, R.; Schimmel, T.; Levron, J.; Willadsen, S. Birth of infant after transfer of anucleate donor oocyte cytoplasm into recipient eggs. Lancet 1997, 350, 186–187. [Google Scholar] [CrossRef]
  44. Cohen, J.; Scott, R.; Alikani, M.; Schimmel, T.; Munné, S.; Levron, J.; Wu, L.; Brenner, C.; Warner, C.; Willadsen, S. Ooplasmic transfer in mature human oocytes. Mol. Hum. Reprod. 1998, 4, 269–280. [Google Scholar] [CrossRef] [PubMed]
  45. Tesarik, J.; Nagy, Z.P.; Mendoza, C.; Greco, E. Chemically and mechanically induced membrane fusion: Non-activating methods for nuclear transfer in mature human oocytes. Hum. Reprod. 2000, 15, 1149–1154. [Google Scholar] [CrossRef] [Green Version]
  46. Tesarik, J. Forty years of in vitro fertilisation: A history of continuous expansion. In 40 Years after In Vitro Fertilisation: State of the Art and New Challenges; Tesarik, J., Ed.; Cambridge Scholars Publishing: Newcastle upon Tyne, UK, 2019; pp. 1–24. [Google Scholar]
  47. Malter, H.E. Improving oocytes and embryos? Cytoplasmic manipulation in human reproduction. In 40 Years after In Vitro Fertilisation: State of the Art and New Challenges; Tesarik, J., Ed.; Cambridge Scholars Publishing: Newcastle upon Tyne, UK, 2019; pp. 234–268. [Google Scholar]
  48. Tesarik, J.; Kopecný, V.; Plachot, M.; Mandelbaum, J. Activation of nucleolar and extranucleolar RNA synthesis and changes in the ribosomal content of human embryos developing in vitro. J. Reprod. Fertil. 1986, 78, 463–470. [Google Scholar] [CrossRef] [Green Version]
  49. Braude, P.; Bolton, V.; Moore, S. Human gene expression first occurs between the four- and eight-cell stages of preimplantation development. Nature 1988, 332, 459–461. [Google Scholar] [CrossRef]
  50. Tesarik, J.; Kopecný, V.; Plachot, M.; Mandelbaum, J. Early morphological signs of embryonic genome expression in human preimplantation development as revealed by quantitative electron microscopy. Dev. Biol. 1988, 128, 15–20. [Google Scholar] [CrossRef]
  51. Tesarik, J. Involvement of oocyte-coded message in cell differentiation control of early human embryos. Development 1989, 105, 317–322. [Google Scholar]
  52. Zhang, J.; Liu, H.; Luo, S.; Lu, Z.; Chávez-Badiola, A.; Liu, Z.; Yang, M.; Merhi, Z.; Silber, S.J.; Munné, S.; et al. Live birth derived from oocyte spindle transfer to prevent mitochondrial disease. Reprod. Biomed. Online 2017, 34, 361–368. [Google Scholar] [CrossRef] [Green Version]
  53. Tesarik, J. Oocyte spindle transfer for prevention of mitochondrial disease: The question of membrane fusion technique. Reprod. Biomed. Online 2017, 35, 432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tesarik, J. Customized assisted reproduction enhancement (CARE) for women with extremely poor ovarian reserve (EPOR). J. Gynecol. Women Health 2017, 3, 555625. [Google Scholar] [CrossRef] [Green Version]
  55. Mendoza-Tesarik, R.; Tesarik, J. Usefulness of individualized FSH, LH and GH dosing in ovarian stimulation of women with low ovarian reserve. Hum. Reprod. 2018, 33, 981–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Overview of the principal nuclear genes with a known role in the protection of the ovaries against aging 1.
Table 1. Overview of the principal nuclear genes with a known role in the protection of the ovaries against aging 1.
GeneLocationFunction
WNT41p36.23-p35.1Female sex determination and differentiation
FIGLA2p13.3Primordial follicle and zona pellucida formation
NOBOX7q35Transition from primordial to growing follicles
FOXO36q21Transition from primordial to growing follicles
PTEN10q23.3Transition from primordial to growing follicles
FSHR2p21-p16Hormone-dependent phase of follicular growth
GPR31p36.1-p35Maintenance of meiotic arrest until the LH surge
MSH41p31DNA mismatch repair during meiotic recombination
MSH56p21.3DNA mismatch repair during meiotic recombination
PGRMC1Xq22-q24Apoptosis of ovarian cells
FOXO113q14.1Granulosa cell function
DMC122q13.1Repair of DNA damage during meiotic divisions
1 See the main text for the full name of each gene. Full review of the genes can be found in references [4,8,9].
Table 2. Agents that can be used to treat the consequences of physiological or premature ovarian aging.
Table 2. Agents that can be used to treat the consequences of physiological or premature ovarian aging.
AgentAdministrationMechanisms of ActionReferences
GHSubcutaneousActivation of cell-signaling pathways acting[26,27,28,29,30,31,32]
against oxidative stress
Possible activation of DNA damage repair
MelatoninOralDirect antioxidant[33,34,35,36]
Indirect antioxidant (signaling pathway modulator)
Anti-inflammatory agent
Immunomodulator
Coenzyme Q10OralDirect antioxidant[37,38,39]
Vitamin COralDirect antioxidant[40]
Vitamin EOralDirect antioxidant[40]
Folic acidOralDirect antioxidant[40]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tesarik, J.; Galán-Lázaro, M.; Mendoza-Tesarik, R. Ovarian Aging: Molecular Mechanisms and Medical Management. Int. J. Mol. Sci. 2021, 22, 1371. https://doi.org/10.3390/ijms22031371

AMA Style

Tesarik J, Galán-Lázaro M, Mendoza-Tesarik R. Ovarian Aging: Molecular Mechanisms and Medical Management. International Journal of Molecular Sciences. 2021; 22(3):1371. https://doi.org/10.3390/ijms22031371

Chicago/Turabian Style

Tesarik, Jan, Maribel Galán-Lázaro, and Raquel Mendoza-Tesarik. 2021. "Ovarian Aging: Molecular Mechanisms and Medical Management" International Journal of Molecular Sciences 22, no. 3: 1371. https://doi.org/10.3390/ijms22031371

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop