Epigenetics, ovarian cell plasticity, and platelet-rich plasma: Mechanistic theories

in Reproduction and Fertility
Authors:
E Scott Sills Office for Reproductive Research, Center for Advanced Genetics/FertiGen, San Clemente, California, USA
Regenerative Biology Group, Fertility Reserve Bank San Clemente, California, USA

Search for other papers by E Scott Sills in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0001-7334-1031
and
Samuel H Wood Gen 5 Fertility Center, San Diego, California, USA

Search for other papers by Samuel H Wood in
Current site
Google Scholar
PubMed
Close

Correspondence should be addressed to E S Sills: ess@prp.md
Open access
Sign up for journal news

Ovarian platelet-rich plasma (PRP) is claimed to restore the fertility potential by improving reserve, an effect perhaps mediated epigenetically by platelet-discharged regulatory elements rather than gonadotropin-activated G-protein coupled receptors, as with stimulated in vitro fertilization (IVF). The finding that fresh activated platelet releasate includes factors able to promote developmental signaling networks necessary to enable cell pluripotency tends to support this theory. The mechanistic uncertainty of intraovarian PRP notwithstanding, at least two other major challenges confront this controversial intervention. The first challenge is to clarify how perimenopausal ovarian function is reset to levels consistent with ovulation. Perhaps a less obvious secondary problem is to confine this renewal such that any induced recalibration of cellular plasticity is kept within acceptable physiologic bounds. Thus, any ‘drive’ to ovarian rejuvenation must incorporate both accelerator and brake. Ovarian aging may be best viewed as a safeguard against pathologic overgrowth, where senescence operates as an evolved tumor-suppression response. While most ovary cells reach the close of their metabolic life span with low risk for hypertrophy, enhanced lysosomal activity and the proinflammatory ‘senescence-associated secretory phenotype’ usually offsets this advantage over time. But is recovery of ovarian fitness possible, even if only briefly prior to IVF? Alterations in gap junctions, bio-conductive features, and modulation of gene regulatory networks after PRP use in other tissues are discussed here alongside early data reported from reproductive medicine.

Abstract

Ovarian platelet-rich plasma (PRP) is claimed to restore the fertility potential by improving reserve, an effect perhaps mediated epigenetically by platelet-discharged regulatory elements rather than gonadotropin-activated G-protein coupled receptors, as with stimulated in vitro fertilization (IVF). The finding that fresh activated platelet releasate includes factors able to promote developmental signaling networks necessary to enable cell pluripotency tends to support this theory. The mechanistic uncertainty of intraovarian PRP notwithstanding, at least two other major challenges confront this controversial intervention. The first challenge is to clarify how perimenopausal ovarian function is reset to levels consistent with ovulation. Perhaps a less obvious secondary problem is to confine this renewal such that any induced recalibration of cellular plasticity is kept within acceptable physiologic bounds. Thus, any ‘drive’ to ovarian rejuvenation must incorporate both accelerator and brake. Ovarian aging may be best viewed as a safeguard against pathologic overgrowth, where senescence operates as an evolved tumor-suppression response. While most ovary cells reach the close of their metabolic life span with low risk for hypertrophy, enhanced lysosomal activity and the proinflammatory ‘senescence-associated secretory phenotype’ usually offsets this advantage over time. But is recovery of ovarian fitness possible, even if only briefly prior to IVF? Alterations in gap junctions, bio-conductive features, and modulation of gene regulatory networks after PRP use in other tissues are discussed here alongside early data reported from reproductive medicine.

Lay summary

Platelet-rich plasma (PRP) is a naturally occurring blood product which has been used to accelerate would healing and correct local aging processes. Recently, PRP has been suggested as an experimental treatment for ‘ovarian rejuvenation’ by injection into older or non-responsive ovaries. This matters because, for now, hormone replacement and in vitro fertilization/egg donation are the only available clinical treatment options for menopause and infertility. Although the gradual loss of ovarian function with increasing age is expected, it might be modified by intraovarian PRP. Indeed, return of menstruation, ovulation, and healthy term deliveries reported after ovarian PRP have moved this treatment beyond the proof-of-concept stage. But how does it actually work, and how might it be validated? Although prior study has focused on platelet cytokine signaling and ovarian stem cells, this proposal discusses new theories for alternative pathways likely to reset the ovarian biological clock.

Introduction

Among the growing list of inputs necessary for folliculogenesis, transforming growth factor-beta 1, growth differentiation factor-9 (GDF-9), vascular endothelial growth factor (VEGF), and insulin-like growth factor-1 (IGF-1) are probably the best known. Platelet-derived growth factor and bone morphogenic proteins also appear central to cell migration, vascular support, and general ovarian function (Sills & Wood 2019). It has been shown that when specified cell exomes releasing these substances are administered near undifferentiated oocyte stem precursors, in an experimental murine ovarian insufficiency model follicle development and restoration of reproductive capacity occur (Li et al. 2021). Likewise, direct injection of activated platelet-rich plasma (PRP) or its derived, condensed plasma cytokines into older ovarian tissue of infertility patients may recover menses, evoke spontaneous ovulation, and enable term live birth (Sills et al. 2018, 2019). When PRP is activated by calcium, this causes discharge of highly purified unbound growth factors along with exosomes. In contrast, insertion of resting (non-activated) platelets for intraovarian use may reduce or even negate the post-treatment response (Rickers & Sills 2022). Since platelet exosome traffic typically accounts for <0.3% of the total protein released in activated PRP (Saumell-Esnaola et al. 2022), this probably adds little to the overall clinical response. This effect (H) of Ca2+ on efflux has been estimated to describe vesicular flow and cytokine moieties leaving activated platelets, as follows:
article image

where m1−m2 is the difference in mean, and s.d.(p) is the pooled/weighted s.d. (Hedges 1981, Saumell-Esnaola et al. 2022). Modeling the microparticle dynamic entangled with activation is useful despite the biochemical composition of platelet releasate being, for now, only partially known. The current work places the processes, receptors, pathways, or combinations of these routes into a theoretical framework consistent with downstream biological observations after autologous fresh intraovarian PRP use.

PRP – ovaria in aeternum?

In an experimental mammal bone marrow-derived mesenchymal stem cell model, investigators from Jiangsu University and Harvard Medical School (Zhang et al. 2020) found that PRP exposure caused the upregulation of key pluripotency genes such as Oct4, Sox2, Sall4, and Nanog. These derivatives show overlap with the set of four genes (c-Myc, Oct3/4, Klf4, and Sox2) which can put somatic cells back on a pluripotent lineage (Takahashi & Yamanaka 2006). Notably, researchers also reported reduced post-PRP expression of β-galactosidase – a known senescence biomarker after PRP use (Zhang et al. 2020).

The reason Oct4 is well placed as a leading contributor to ovarian remodeling is that human gonadal (testicular) cells express Oct4 (and c-KIT) after in vitro culture with PRP (Khadivi et al. 2020). It is plausible that a parallel response occurs in adult ovarian tissues following intraovarian activated platelet-derived cytokines (Sills & Wood 2022). Mao et al. (2014) tested the hypothesis that granulosa cells might display pluripotency by a novel shortcut using fewer regulatory factors. Indeed, they succeeded in generating this at high efficiency with only two factors, Oct4 and Sox2 (Mao et al. 2014). The only known factors that might substitute for Oct4 in human cellular reprogramming are a synthetic fusion protein of the lineage specifier GATA3-VP16 (but with much lower yield than Oct4, mir302/367, or mir200c/302/369) (Miyoshi et al. 2011, Montserrat et al. 2013).

The lack of evidence for a naturally occurring transcription factor capable of promoting Oct4 suggests that there are some uncharacterized factors which are essential for proper reprogramming (Mai et al. 2018), a conclusion which encourages research focusing on numerous still-incompletely defined PRP components.

PRP and DNA methylation

Epigenetic processes modulate gene expression without changing the basic DNA sequence; specific genes are silenced via transcriptional repression during differentiation and growth (Smith & Meissner 2013). This is done by adding or removing chromatin modifications by posttranslational modification (Bannister & Kouzarides 2011). Examples include acetylation, methylation, or phosphorylation which encounter protein complexes to direct gene expression (Li & Li 2021). In mammals, this steric adjustment usually involves methylation at cytosine or adenine nucleobases. For cytosine, its conversion to 5-methylcytosine (5mC) usually yields repression, while adenine becoming N6-methyladenine is associated with gene amplification (Yasmin et al. 2015, Deniz et al. 2019). Epigenetic reprogramming in mammals is accomplished in two phases to reset the pluripotent potential by clearing away prior patterns governing cell lineage. One of these steps occurs somatically, while the second operates in the germline. In this way, all previous DNA methylation is erased to establish a new, de novo pattern for restoration of the epigenome (Sendžikaitė & Kelsey 2019, Greenberg & Bourc'his 2019).

If pregnancy were established by a young mother, such offspring would derive from an oocyte typified by a partially open chromatin structure (Schächner et al. 2022). However, if conception were delayed much beyond 35 years of age that oocyte will be substantially different, characterized by sharply reduced 5mC in the egg genome. This constrains repression of transposable elements (Yoder et al. 1997), placing insertional mutations within coding and/or regulatory sequences to accumulate as net genomic instability (López-Otín et al. 2013). Indeed, the mature human oocyte released at 30 years of age already has at least 1.5× more transposable elements than at 20 years of age (Sturm & Vellai 2022). This mutagenesis tracks to senescence and, over time, leads to loss of the affected cell (Sturm et al. 2017, Gorbunova et al. 2021).

An intriguing feature common among apparently ageless and potentially immortal eukaryotic systems (i.e. cnidaria, planarian flatworm) is ‘P‐element‐induced wimpy testis in Drosophila’ – Piwi‐interacting noncoding RNA (Piwi-piRNA) pathway (Sturm et al. 2017). This gene regulator squelches mobile transposable elements which move from one locus to another, resulting in mutation. These 'jumping genes' amplify with old age, underscoring their role in genetic information decay with advanced age (Sturm et al. 2017). In contrast, planarian flatworms and freshwater hydra enjoy seemingly unlimited self-renewal by embodying components of this Piwi‐piRNA mechanism (Petralia et al. 2014).

In adult human ovarian tissue, characterization of Piwi‐piRNA is preliminary, although basic aspects have already been discovered in testes (Moody et al. 2020). Present in hematopoietic stem cells and certain malignancies, Piwi-piRNA engages to produce essentially unbounded proliferation and renewal. Notably, where the Piwi‐piRNA pathway is inactive, significant intron error accrues (Sturm et al. 2017). Once dismissed as ‘junk DNA’, introns and their alterations are associated with normal aging and associated pathologies (Huang et al. 2012, Kreiling et al. 2017). If retrotransposon dynamics rise exponentially with age, the uptick in copy number would align with the mortality pattern observed in many animal species (Sturm et al. 2017).

Genome-wide analysis of rat DNA methylation patterns for genes relevant to hormone control, glutamate signaling, and melatonin and circadian pathways suggests that menopause onset is under epigenetic control (Bacon et al. 2019). Moreover, histone and methylation changes occur across the perimenopause transition, suggesting epigenetic reorganization begins well ahead of menopause typified by loss of plasticity potential (Yin et al. 2015, Bacon et al. 2019). Activity of DNA methyltransferases is at least partially under the control of miRNAs, supplied as a constituent of activated PRP (see Fig. 1). These neuroendocrine studies encourage ongoing parallel investigations in a human ovary setting.

Figure 1
Figure 1

Fresh autologous platelet-rich plasma (p) is activated (green) to produce p’, enabling platelet releaseate with at least three constituents – miRNAs, multiple cytokines/growth factors, as well as gene regulators targeting undifferentiated oocyte precursors. Via single-needle puncture, p’ is injected into ovarian stroma (A) and subcapsular segments (red). Serum AMH is periodically monitored [3] using a uniform assay for documentation of ovarian reserve response (B), leading to follicular development (purple) and occasional spontaneous ovulation (C).

Citation: Reproduction and Fertility 3, 4; 10.1530/RAF-22-0078

Measuring menopause and how to reset it

As menopause nears, it brings a maternal refractoriness to gonadotropins in metabolically depleted ovaries which renders oocyte recruitment during IVF essentially futile. The static count for resting/non-growing follicles declines sharply with age, and this loss has been estimated within the entire ovary field (Hansen et al. 2008). Setting mean menopause age at 51 years, less than 800 resting follicles are predicted to remain in both ovaries at menopause, with 95% CI for age of menopause between 41.9 and 57.8 years:

log (n) = (–0.00019) × ay 2.452 + 5.717

where ay = age in years. Here, no sudden exhaustion is described but rather a constantly increasing decay rate. The model not only agrees well with observed ages of menopause but shows that a considerable percentage of follicle decline cannot be explained by age alone. Even when limited to primordial follicles, the model is still a better fit to the data than any previous model (Hansen et al. 2008). The accompanying perimenopausal neuroendocrine milieu characterized by chronic follicle stimulating hormone (FSH) elevation would seem to undercut the clinical rationale for administering still more (high dose and synthetic) gonadotropins, unless recapture of gonadal sensitivity could somehow be produced first.

If ovarian stem-like cells are resident in the subcapsular layer even in older/menopausal ovaries (Sills & Wood 2019), these important targets would remain indifferent to conventional IVF stimulation until proper recruitment and development signals are communicated first. As reported previously, interleukin-7 (IL-7) starts a signaling cascade favoring downstream release of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and IL-10, which then regulate genes for oocyte maturation (Zhang et al. 2020, Sills & Wood 2022). In contrast, GDF-15, IFN-γ, insulin-like growth factor-binding protein-2 (IGFBP-2), IGFBP-7, IL-15, IL-1β, IL-17A, IL-8, monocyte chemoattractant protein-1 (MCP-1), tissue inhibitors of metalloproteinase-2, TNF-α, VEGF-A, and interferon-inducible protein-10 have been associated with aging ovarian status (Shin et al. 2022). When the oocyte-specific homeobox gene (Nobox) is muted, the postnatal oocyte pool is severely curtailed in mice by interrupting the transition from primordial to growing follicles. Echoing features seen in human menopause and low ovarian reserve, murine follicles are overtaken by fibrosis if Nobox is absent. Indeed, Rajkovic et al. were among the first to show how Nobox knockout causes sharp downregulation of Oct4 and Gdf9, genes known to be preferentially expressed in murine oocytes (Rajkovic et al. 2004). These pluripotency markers (with Sox2, Sall4, and Nanog) are amplified following local intraovarian PRP dosing (Zhang et al. 2020).

Interestingly, recent cross-species study reported on uterine lavage using human PRP to treat mouse intracavitary uterine synechiae (Kim et al. 2022), where significantly increased STAT3 phosphorylation (a critical transcription factor in tissue regeneration) was observed with upregulated Mt2-mmp, Lox, and Adm, all mediators of tissue remodeling. Cell proliferation, maintenance of genome stability, and metabolic regulation are all influenced by sirtuins (Nielsen et al. 2022), which are NAD-dependent deacetylases. Investigators at Huazhong Science & Technology University discovered that SIRT1, SIRT3, and SIRT6 are closely related to ovarian reserve and may serve as proxy markers of ovarian aging (Zhang et al. 2016). Researchers from Jilin University later carried this forward to show activated PRP accelerates differentiation and viability of specific cell types via SIRT1 (Xu et al. 2021). Recording how autologous activated PRP affects the SIRT group specifically in the adult human ovary is planned.

Cellular conductance and bioelectric considerations

Specialized protein channels are required to facilitate fertilization, immune response, and cellular bioelectric status, as free ion migration is checked by hydrophobic lipid membranes (Gadsby et al. 2009). For human luteinized granulosa cells, potassium and sodium channels are homologous with those in neuroendocrine cells of adrenal and thyroid C cells (Klugbauer et al. 1995, Bulling et al. 1999). How these ion gates change within ovaries during aging is being studied, but manipulation of cell cycle regulators via redox inputs and voltage gradients has been shown to influence rejuvenation elsewhere (Durant et al. 2017, Weng et al. 2021, Bakalova et al. 2022).

For example, intercellular gap junction manipulations can adjust cell polarity and potentiate signal transmission capable of dramatic reprogramming of biological morphologies (Bischof et al. 2020, Cervera et al. 2021). Phenotypic consequences for senescent ovary systems have not yet been fully described but are expected to mimic previously reported regeneration to reestablish function, reset a distributed goal state, and correct large-scale anatomies and/or functions from initial conditions (Pezzulo et al. 2021). How is this possible? Ongoing transcriptional systems are highly plastic and retain much potential for remodeling, yet little is known about this ‘bioelectrostat’ which may be miscalibrated either too high or too low. Disordered transmembrane ion traffic is well researched in cardiac disease, schizophrenia, bipolar disorder, and Alzheimer's disease (Berridge 2012) but not yet in ovarian senescence.

Also of importance are cellular water levels, which fluctuate over time as with electrolytes. In female reproduction, aquaporins manage water transport at intraluminal, interstitial, and capillary levels, largely under progesterone direction (Kordowitzki et al. 2020). Previous work has shown that at least 11 aquaporin isoforms operate in the female reproductive tract of various mammals, including human. The first aquaporin in the female reproductive system was confirmed by isolating cDNA encoding for a water channel generated from (human) uterus where cloned cDNA showed close homology (99.8%) with the 28 kDa human erythrocyte CHIP28 (Agre et al. 1993). While the function of transmembrane channels is subject to resting voltage potentials (Bulling et al. 1999), how platelet (PLT) cytokines/PRP may impact ovarian cell membrane portal dynamics is only now being investigated. This should extend the fibroblast work ofSassoli et al. (2022) who were the first to observe a mechanism involving gap junction currents as operational targets for selected platelet cytokines.

Conclusions

One reason for age-associated ovarian disability could be due to gradual maladaptive epigenetic noise which gathers over time to dysregulate gene expression, culminating in the familiar ‘poor ovarian reserve’ picture commonly seen in clinical IVF practice. As human ovarian function declines with age, resetting plasticity via cell development switches actuated by external inputs might be within reach – as we propose here. This is less revolutionary than at first it may seem. The chronicle of biological science sometimes skips the chapter on phage λ lytic cycle regulation, now an obscure example of plasticity mapping (Ptashne 1992, Sommer 2020). Documenting how autologous PRP could supply the essential stimulus to reset ovarian plasticity is the topic of current studies.

While the subject of how ovary cells cease their growth once an optimal point has been reached frames a central tenet of developmental biology, it remains incompletely understood. For plants, root growth cessation requires action of an auxin influx carrier, AUX1 (Liu et al. 2022). For animals, miRNAs regulate various biological processes including apoptosis and proliferation (Gareev et al. 2021). Because at least 60% of human protein-coding genes are directly regulated by miRNAs (Dexheimer & Cochella 2020) and most circulating miRNAs are of platelet origin (Czajka et al. 2021), the behavior of this PRP component within human ovary is an attractive topic for research (Sills & Wood 2022).

Understandably, there is hope that such searches for alternatives to oocyte donation, IVF, synthetic hormones, or gonadotropins will gain validation. Resumed menses after menopause or deliveries for infertility patients after intraovarian PRP have drawn scrutiny on this experimental procedure. While case reports and small series are encouraging, strong evidence to support its general application is missing (Kheil et al. 2022). But to claim the topic is unimportant to women’s reproductive health seems incorrect, as a recent survey of world output on intraovarian PRP research (Sills & Wood 2021) was soon eclipsed by new datasets published from leading centers. For example, Australian experts reported on 20 consecutive IVF patients with severely reduced reserve to compare outcomes before vs after intraovarian PRP (Tremellen & Pacella-Ince 2022). Following non-activated PRP administered via 19G needle performed under sedation, IVF was completed 2––3 months later. Among six patients achieving pregnancy, the mean age was >40 years; there were no losses by 20 weeks gestation and two have since delivered (Tremellen & Pacella-Ince 2022). A protocol from New York included 20G needle injection of non-activated PRP with up to 12 punctures per ovary, followed by stimulated IVF cycles after ~1 month (Barad et al. 2022). Close endocrine monitoring of 80 cases revealed no clinically significant effects on ovarian function, but two deliveries occurred (both patients’ age = 40 years) during an impressive 1-year follow-up interval (Barad et al. 2022). Perhaps the largest single-center intraovarian PRP experience to date (n = 510) originates from Istanbul (Cakiroglu et al. 2022) where autologous PRP (activation unspecified) was injected via 17G needle under sedation. The mean patient age in the series was 40.3 years, with post-PRP pregnancy and live birth rates of 20.5 and 12.9%, respectively (Cakiroglu et al. 2022).

These outcomes – regardless of which IVF calendar or what needle is used for PRP injection – suggest that such details may be subsidiary so long as clinics follow protocols most familiar to them (Sills et al. 2018). Descriptive reports can add to the PRP knowledge fund, as interested researchers gain a better understanding of methods and techniques for ‘ovarian rejuvenation’.

Declaration of interest

ESS has been awarded U.S. Trademark #88505430 for treatment of female hormone and fertility enhancement that utilizes a specified method of intraovarian injection of autologous PRP.

Funding

None.

Author contribution statement

ESS developed the research plans and initial drafts; ESS and SHW reviewed the literature and edited the work. Both authors read and approved the final manuscript.

References

  • Agre P, Preston GM, Smith BL, Jung JS, Raina S, Moon C, Guggino WB & Nielsen S 1993 Aquaporin CHIP: the archetypal molecular water channel. American Journal of Physiology 265 F463F476. (https://doi.org/10.1152/ajprenal.1993.265.4.F463)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bacon ER, Mishra A, Wang Y, Desai MK, Yin F & Brinton RD 2019 Neuroendocrine aging precedes perimenopause and is regulated by DNA methylation. Neurobiology of Aging 74 213224. (https://doi.org/10.1016/j.neurobiolaging.2018.09.029)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bakalova R, Aoki I, Zhelev Z & Higashi T 2022 Cellular redox imbalance on the crossroad between mitochondrial dysfunction, senescence, and proliferation. Redox Biology 53 102337. (https://doi.org/10.1016/j.redox.2022.102337)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bannister AJ & Kouzarides T 2011 Regulation of chromatin by histone modifications. Cell Research 21 381395. (https://doi.org/10.1038/cr.2011.22)

  • Barad DH, Albertini DF, Molinari E & Gleicher N 2022 Preliminary report of intraovarian injections of autologous platelet-rich plasma (PRP) in extremely poor prognosis patients with only oocyte donation as alternative: a prospective cohort study. Human Reproduction Open 2022 hoac027. (https://doi.org/10.1093/hropen/hoac027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Berridge MJ 2012 Calcium signalling remodelling and disease. Biochemical Society Transactions 40 297309. (https://doi.org/10.1042/BST20110766)

  • Bischof J, Day ME, Miller KA, LaPalme JV & Levin M 2020 Nervous system and tissue polarity dynamically adapt to new morphologies in planaria. Developmental Biology 467 5165. (https://doi.org/10.1016/j.ydbio.2020.08.009)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulling A, Brucker C, Berg U, Gratzl M & Mayerhofer A 1999 Identification of voltage-activated Na+ and K+ channels in human steroid-secreting ovarian cells. Annals of the New York Academy of Sciences 868 7779. (https://doi.org/10.1111/j.1749-6632.1999.tb11275.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cakiroglu Y, Yuceturk A, Karaosmanoglu O, Kopuk SY, Korun ZEU, Herlihy N, Scott RT, Tiras B & Seli E 2022 Ovarian reserve parameters and IVF outcomes in 510 women with poor ovarian response (POR) treated with intraovarian injection of autologous platelet rich plasma (PRP) Aging (Albany NY) 14 25132523. (https://doi.org/10.18632/aging.203972)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cervera J, Levin M & Mafe S 2021 Morphology changes induced by intercellular gap junction blocking: A reaction-diffusion mechanism. Biosystems 209 104511. (https://doi.org/10.1016/j.biosystems.2021.104511)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Czajka P, Fitas A, Jakubik D, Eyileten C, Gasecka A, Wicik Z, Siller-Matula JM, Filipiak KJ & Postula M 2021 MicroRNA as potential biomarkers of platelet function on antiplatelet therapy: a review. Frontiers in Physiology 12 652579. (https://doi.org/10.3389/fphys.2021.652579)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deniz Ö, Frost JM & Branco MR 2019 Regulation of transposable elements by DNA modifications. Nature Reviews. Genetics 20 417431. (https://doi.org/10.1038/s41576-019-0106-6)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dexheimer PJ & Cochella L 2020 MicroRNAs: from mechanism to organism. Frontiers in Cell and Developmental Biology 8 409. (https://doi.org/10.3389/fcell.2020.00409)

  • Durant F, Morokuma J, Fields C, Williams K, Adams DS & Levin M 2017 Long-term, stochastic editing of regenerative anatomy via targeting endogenous bioelectric gradients. Biophysical Journal 112 22312243. (https://doi.org/10.1016/j.bpj.2017.04.011)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gadsby DC 2009 Ion channels versus ion pumps: the principal difference, in principle. Nature Reviews. Molecular Cell Biology 10 344352. (https://doi.org/10.1038/nrm2668)

  • Gareev I, Beylerli O, Liang Y, Xiang H, Liu C, Xu X, Yuan C, Ahmad A & Yang G 2021 The role of microRNAs in therapeutic resistance of malignant primary brain tumors. Frontiers in Cell and Developmental Biology 9 740303. (https://doi.org/10.3389/fcell.2021.740303)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gorbunova V, Seluanov A, Mita P, McKerrow W, Fenyö D, Boeke JD, Linker SB, Gage FH, Kreiling JA & Petrashen AP et al.2021 The role of retrotransposable elements in ageing and age-associated diseases. Nature 596 4353. (https://doi.org/10.1038/s41586-021-03542-y)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greenberg MVC & Bourc'his D 2019 The diverse roles of DNA methylation in mammalian development and disease. Nature Reviews. Molecular Cell Biology 20 590607. (https://doi.org/10.1038/s41580-019-0159-6)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hansen KR, Knowlton NS, Thyer AC, Charleston JS, Soules MR & Klein NA 2008 A new model of reproductive aging: the decline in ovarian non-growing follicle number from birth to menopause. Human Reproduction 23 699708. (https://doi.org/10.1093/humrep/dem408)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hedges LV 1981 Distribution theory for Glass’s estimator of effect size and related estimators. Journal of Educational Statistics 6 107128. (https://doi.org/10.3102/10769986006002107)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang CR, Burns KH & Boeke JD 2012 Active transposition in genomes. Annual Review of Genetics 46 651675. (https://doi.org/10.1146/annurev-genet-110711-155616)

  • Khadivi F, Koruji M, Akbari M, Jabari A, Talebi A, Ashouri Movassagh S, Panahi Boroujeni A, Feizollahi N, Nikmahzar A & Pourahmadi M et al.2020 Application of platelet-rich plasma (PRP) improves self-renewal of human spermatogonial stem cells in two-dimensional and three-dimensional culture systems. Acta Histochemica 122 151627. (https://doi.org/10.1016/j.acthis.2020.151627)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kheil MH, Bahsoun R & Sharara FI 2022 Platelet-rich plasma: inconclusive evidence of reproductive outcomes in menopausal women. Journal of Assisted Reproduction and Genetics 39 19871991. (https://doi.org/10.1007/s10815-022-02554-5)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim MK, Yoon JA, Yoon SY, Park M, Lee WS, Lyu SW & Song H 2022 Human platelet-rich plasma facilitates angiogenesis to restore impaired uterine environments with Asherman's syndrome for embryo implantation and following pregnancy in mice. Cells 11 1549. (https://doi.org/10.3390/cells11091549)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Klugbauer N, Lacinova L, Flockerzi V & Hofmann F 1995 Structure and functional expression of a new member of the tetrodotoxin-sensitive voltage-activated sodium channel family from human neuroendocrine cells. EMBO Journal 14 10841090. (https://doi.org/10.1002/j.1460-2075.1995.tb07091.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kordowitzki P, Kranc W, Bryl R, Kempisty B, Skowronska A & Skowronski MT 2020 The relevance of aquaporins for the physiology, pathology, and aging of the female reproductive system in mammals. Cells 9 E2570. (https://doi.org/10.3390/cells9122570)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kreiling JA, Jones BC, Wood JG, De Cecco M, Criscione SW & Neretti N et al.2017 Contribution of retrotransposable elements to aging. In Human Retrotransposons in Health and Disease. Cham, CH: Springer. Cristofari GEd., pp. 297321. (https://doi.org/10.1007/978-3-319-48344-3_13)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li X & Li XD 2021 Integrative chemical biology approaches to deciphering the histone code: a problem-driven journey. Accounts of Chemical Research 54 37343747. (https://doi.org/10.1021/acs.accounts.1c00463)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Z, Zhang M, Zheng J, Tian Y, Zhang H, Tan Y, Li Q, Zhang J & Huang X 2021 Human umbilical cord mesenchymal stem cell-derived exosomes improve ovarian function and proliferation of premature ovarian insufficiency by regulating the Hippo signaling pathway. Frontiers in Endocrinology (Lausanne) 12 711902. (https://doi.org/10.3389/fendo.2021.711902)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu S, Strauss S, Adibi M, Mosca G, Yoshida S, Dello Ioio R, Runions A, Andersen TG, Grossmann G & Huijser P et al.2022 Cytokinin promotes growth cessation in the Arabidopsis root. Current Biology 32 19741985.e3. (https://doi.org/10.1016/j.cub.2022.03.019)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • López-Otín C, Blasco MA, Partridge L, Serrano M & Kroemer G 2013 The hallmarks of aging. Cell 153 11941217. (https://doi.org/10.1016/j.cell.2013.05.039)

  • Mai T, Markov GJ, Brady JJ, Palla A, Zeng H, Sebastiano V & Blau HM 2018 NKX3-1 is required for induced pluripotent stem cell reprogramming and can replace OCT4 in mouse and human iPSC induction. Nature Cell Biology 20 900908. (https://doi.org/10.1038/s41556-018-0136-x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mao J, Zhang Q, Ye X, Liu K & Liu L 2014 Efficient induction of pluripotent stem cells from granulosa cells by Oct4 and Sox2. Stem Cells and Development 23 779789. (https://doi.org/10.1089/scd.2013.0325)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Miyoshi N, Ishii H, Nagano H, Haraguchi N, Dewi DL, Kano Y, Nishikawa S, Tanemura M, Mimori K & Tanaka F et al.2011 Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell 8 633638. (https://doi.org/10.1016/j.stem.2011.05.001)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Montserrat N, Nivet E, Sancho-Martinez I, Hishida T, Kumar S, Miquel L, Cortina C, Hishida Y, Xia Y & Esteban CR et al.2013 Reprogramming of human fibroblasts to pluripotency with lineage specifiers. Cell Stem Cell 13 341350. (https://doi.org/10.1016/j.stem.2013.06.019)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moody SC, Wakitani S, Young JC, Western PS & Loveland KL 2020 Evidence that activin A directly modulates early human male germline differentiation status. Reproduction 160 141154. (https://doi.org/10.1530/REP-20-0095)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nielsen JL, Bakula D & Scheibye-Knudsen M 2022 Clinical trials targeting aging. Frontiers in Aging 3 820215. (https://doi.org/10.3389/fragi.2022.820215)

  • Petralia RS, Mattson MP & Yao PJ 2014 Aging and longevity in the simplest animals and the quest for immortality. Ageing Research Reviews 16 6682. (https://doi.org/10.1016/j.arr.2014.05.003)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pezzulo G, LaPalme J, Durant F & Levin M 2021 Bistability of somatic pattern memories: stochastic outcomes in bioelectric circuits underlying regeneration. Philosophical Transactions of the Royal Society of London Series B 376(1821) 20190765. (https://doi.org/10.1098/rstb.2019.0765)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ptashne M 1992 A genetic switch: phage and higher organisms. Cambridge, MA: Blackwell Publishing Scientific & Cell Press, pp. 1192. Available at: http://hdl.handle.net/10822/545304

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rajkovic A, Pangas SA, Ballow D, Suzumori N & Matzuk MM 2004 NOBOX deficiency disrupts early folliculogenesis and oocyte-specific gene expression. Science 305 11571159. (https://doi.org/10.1126/science.1099755)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rickers NS & Sills ES 2022 Is autologous platelet activation the key step in ovarian therapy for fertility recovery and menopause reversal? Taipei: Biomedicine.

  • Sassoli C, Garella R, Chellini F, Tani A, Pavan P, Bambi F, Zecchi-Orlandini S & Squecco R 2022 Platelet-rich plasma affects gap junctional features in myofibroblasts in vitro via vascular endothelial growth factor (VEGF)-A/VEGF receptor. Experimental Physiology 107 106121. (https://doi.org/10.1113/EP090052)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Saumell-Esnaola M, Delgado D, García Del Caño G, Beitia M, Sallés J, González-Burguera I, Sánchez P, López de Jesús M, Barrondo S & Sánchez M 2022 Isolation of platelet-derived exosomes from human platelet-rich plasma: biochemical and morphological characterization. International Journal of Molecular Sciences 23 2861. (https://doi.org/10.3390/ijms23052861)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schächner C, Merkl PE, Pilsl M, Schwank K, Hergert K, Kruse S, Milkereit P, Tschochner H & Griesenbeck J 2022 Establishment and maintenance of open ribosomal RNA gene chromatin states in eukaryotes. Methods in Molecular Biology 2533 2538. (https://doi.org/10.1007/978-1-0716-2501-9_2)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sendžikaitė G & Kelsey G 2019 The role and mechanisms of DNA methylation in the oocyte. Essays in Biochemistry 63 691705. (https://doi.org/10.1042/EBC20190043)

  • Shin JW, Lee E, Han S, Choe SA & Jeon OH 2022 Plasma proteomic signature of cellular senescence and markers of biological aging among postmenopausal women. Rejuvenation Research 25 141148. (https://doi.org/10.1089/rej.2022.0024)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES, Li X, Rickers NS, Wood SH & Palermo GD 2019 Metabolic and neurobehavioral response following intraovarian administration of autologous activated platelet-rich plasma: first qualitative data. Neuro Endocrinology Letters 39 427433. Available at: https://www.nel.edu/userfiles/articlesnew/1553033179_39_6_sills_427-pdf.pdf

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES, Rickers NS, Li X & Palermo GD 2018 First data on in vitro fertilization and blastocyst formation after intraovarian injection of calcium gluconate-activated autologous platelet rich plasma. Gynecological Endocrinology 34 756760. (https://doi.org/10.1080/09513590.2018.1445219)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES, Rickers NS & Wood SH 2020 Intraovarian insertion of autologous platelet growth factors as cell-free concentrate: Fertility recovery and first unassisted conception with term delivery at age over 40. International Journal of Reproductive Biomedicine 18 10811086. (https://doi.org/10.18502/ijrm.v18i12.8030)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES & Wood SH 2019 Autologous activated platelet-rich plasma injection into adult human ovary tissue: molecular mechanism, analysis, and discussion of reproductive response. Bioscience Reports 39 BSR20190805. (https://doi.org/10.1042/BSR20190805)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES & Wood SH 2021 Progress in human ovarian rejuvenation: Current platelet-rich plasma and condensed cytokine research activity by scope and international origin. Clinical and Experimental Reproductive Medicine 48 311315. (https://doi.org/10.5653/cerm.2021.04651)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES & Wood SH 2022 Growth factors, gene activation, and cell recruitment: from intraovarian condensed platelet cytokines to denovo oocyte development. Journal of Clinical and Translational Research 8 4953. Available at: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8848765/

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Smith ZD & Meissner A 2013 DNA methylation: roles in mammalian development. Nature Reviews. Genetics 14 204220. (https://doi.org/10.1038/nrg3354)

  • Sommer RJ 2020 Phenotypic plasticity: From theory and genetics to current and future challenges. Genetics 215 113. (https://doi.org/10.1534/genetics.120.303163)

  • Sturm Á, Perczel A, Ivics Z & Vellai T 2017 The piwi-piRNA pathway: Road to immortality. Aging Cell 16 906911. (https://doi.org/10.1111/acel.12630)

  • Sturm Á & Vellai T 2022 How does maternal age affect genomic stability in the offspring? Aging Cell 21 e13612. (https://doi.org/10.1111/acel.13612)

  • Takahashi K & Yamanaka S 2006 Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126 663676. (https://doi.org/10.1016/j.cell.2006.07.024)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tremellen K & Pacella-Ince L 2022 An audit of clinical outcomes following ovarian administration of platelet-rich plasma (PRP) in women with severe diminished ovarian reserve. Australian and New Zealand Journal of Obstetrics and Gynaecology 62 767772. (https://doi.org/10.1111/ajo.13578)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Weng HP, Cheng YY, Lee HL, Hsu TY, Chang YT & Shen YA 2021 Enhanced platelet-rich plasma (ePRP) stimulates wound healing through effects on metabolic reprogramming in fibroblasts. International Journal of Molecular Sciences 22 12623. (https://doi.org/10.3390/ijms222312623)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xu Y, Wang X, Liu W & Lu W 2021 Thrombin-activated platelet-rich plasma enhances osteogenic differentiation of human periodontal ligament stem cells by activating SIRT1-mediated autophagy. European Journal of Medical Research 26 105. (https://doi.org/10.1186/s40001-021-00575-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yasmin R, Siraj S, Hassan A, Khan AR, Abbasi R & Ahmad N 2015 Epigenetic regulation of inflammatory cytokines and associated genes in human malignancies. Mediators of Inflammation 2015 201703. (https://doi.org/10.1155/2015/201703)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yin F, Yao J, Sancheti H, Feng T, Melcangi RC, Morgan TE, Finch CE, Pike CJ, Mack WJ & Cadenas E et al.2015 The perimenopausal aging transition in the female rat brain: decline in bioenergetic systems and synaptic plasticity. Neurobiology of Aging 36 22822295. (https://doi.org/10.1016/j.neurobiolaging.2015.03.013)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yoder JA, Walsh CP & Bestor TH 1997 Cytosine methylation and the ecology of intragenomic parasites. Trends in Genetics 13 335340. (https://doi.org/10.1016/s0168-9525(9701181-5)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang J, Fang L, Lu Z, Xiong J, Wu M, Shi L, Luo A & Wang S 2016 Are sirtuins markers of ovarian aging? Gene 575 680686. (https://doi.org/10.1016/j.gene.2015.09.043)

  • Zhang J, Zhang J, Zhang N, Li T, Zhou X, Jia J, Liang Y, Sun X & Chen H 2020 The effects of platelet-rich and platelet-poor plasma on biological characteristics of BM-MSCs in vitro. Analytical Cellular Pathology 2020 8546231. (https://doi.org/10.1155/2020/8546231)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Figure 1

    Fresh autologous platelet-rich plasma (p) is activated (green) to produce p’, enabling platelet releaseate with at least three constituents – miRNAs, multiple cytokines/growth factors, as well as gene regulators targeting undifferentiated oocyte precursors. Via single-needle puncture, p’ is injected into ovarian stroma (A) and subcapsular segments (red). Serum AMH is periodically monitored [3] using a uniform assay for documentation of ovarian reserve response (B), leading to follicular development (purple) and occasional spontaneous ovulation (C).

  • Agre P, Preston GM, Smith BL, Jung JS, Raina S, Moon C, Guggino WB & Nielsen S 1993 Aquaporin CHIP: the archetypal molecular water channel. American Journal of Physiology 265 F463F476. (https://doi.org/10.1152/ajprenal.1993.265.4.F463)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bacon ER, Mishra A, Wang Y, Desai MK, Yin F & Brinton RD 2019 Neuroendocrine aging precedes perimenopause and is regulated by DNA methylation. Neurobiology of Aging 74 213224. (https://doi.org/10.1016/j.neurobiolaging.2018.09.029)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bakalova R, Aoki I, Zhelev Z & Higashi T 2022 Cellular redox imbalance on the crossroad between mitochondrial dysfunction, senescence, and proliferation. Redox Biology 53 102337. (https://doi.org/10.1016/j.redox.2022.102337)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bannister AJ & Kouzarides T 2011 Regulation of chromatin by histone modifications. Cell Research 21 381395. (https://doi.org/10.1038/cr.2011.22)

  • Barad DH, Albertini DF, Molinari E & Gleicher N 2022 Preliminary report of intraovarian injections of autologous platelet-rich plasma (PRP) in extremely poor prognosis patients with only oocyte donation as alternative: a prospective cohort study. Human Reproduction Open 2022 hoac027. (https://doi.org/10.1093/hropen/hoac027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Berridge MJ 2012 Calcium signalling remodelling and disease. Biochemical Society Transactions 40 297309. (https://doi.org/10.1042/BST20110766)

  • Bischof J, Day ME, Miller KA, LaPalme JV & Levin M 2020 Nervous system and tissue polarity dynamically adapt to new morphologies in planaria. Developmental Biology 467 5165. (https://doi.org/10.1016/j.ydbio.2020.08.009)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulling A, Brucker C, Berg U, Gratzl M & Mayerhofer A 1999 Identification of voltage-activated Na+ and K+ channels in human steroid-secreting ovarian cells. Annals of the New York Academy of Sciences 868 7779. (https://doi.org/10.1111/j.1749-6632.1999.tb11275.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cakiroglu Y, Yuceturk A, Karaosmanoglu O, Kopuk SY, Korun ZEU, Herlihy N, Scott RT, Tiras B & Seli E 2022 Ovarian reserve parameters and IVF outcomes in 510 women with poor ovarian response (POR) treated with intraovarian injection of autologous platelet rich plasma (PRP) Aging (Albany NY) 14 25132523. (https://doi.org/10.18632/aging.203972)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cervera J, Levin M & Mafe S 2021 Morphology changes induced by intercellular gap junction blocking: A reaction-diffusion mechanism. Biosystems 209 104511. (https://doi.org/10.1016/j.biosystems.2021.104511)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Czajka P, Fitas A, Jakubik D, Eyileten C, Gasecka A, Wicik Z, Siller-Matula JM, Filipiak KJ & Postula M 2021 MicroRNA as potential biomarkers of platelet function on antiplatelet therapy: a review. Frontiers in Physiology 12 652579. (https://doi.org/10.3389/fphys.2021.652579)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deniz Ö, Frost JM & Branco MR 2019 Regulation of transposable elements by DNA modifications. Nature Reviews. Genetics 20 417431. (https://doi.org/10.1038/s41576-019-0106-6)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dexheimer PJ & Cochella L 2020 MicroRNAs: from mechanism to organism. Frontiers in Cell and Developmental Biology 8 409. (https://doi.org/10.3389/fcell.2020.00409)

  • Durant F, Morokuma J, Fields C, Williams K, Adams DS & Levin M 2017 Long-term, stochastic editing of regenerative anatomy via targeting endogenous bioelectric gradients. Biophysical Journal 112 22312243. (https://doi.org/10.1016/j.bpj.2017.04.011)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gadsby DC 2009 Ion channels versus ion pumps: the principal difference, in principle. Nature Reviews. Molecular Cell Biology 10 344352. (https://doi.org/10.1038/nrm2668)

  • Gareev I, Beylerli O, Liang Y, Xiang H, Liu C, Xu X, Yuan C, Ahmad A & Yang G 2021 The role of microRNAs in therapeutic resistance of malignant primary brain tumors. Frontiers in Cell and Developmental Biology 9 740303. (https://doi.org/10.3389/fcell.2021.740303)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gorbunova V, Seluanov A, Mita P, McKerrow W, Fenyö D, Boeke JD, Linker SB, Gage FH, Kreiling JA & Petrashen AP et al.2021 The role of retrotransposable elements in ageing and age-associated diseases. Nature 596 4353. (https://doi.org/10.1038/s41586-021-03542-y)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greenberg MVC & Bourc'his D 2019 The diverse roles of DNA methylation in mammalian development and disease. Nature Reviews. Molecular Cell Biology 20 590607. (https://doi.org/10.1038/s41580-019-0159-6)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hansen KR, Knowlton NS, Thyer AC, Charleston JS, Soules MR & Klein NA 2008 A new model of reproductive aging: the decline in ovarian non-growing follicle number from birth to menopause. Human Reproduction 23 699708. (https://doi.org/10.1093/humrep/dem408)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hedges LV 1981 Distribution theory for Glass’s estimator of effect size and related estimators. Journal of Educational Statistics 6 107128. (https://doi.org/10.3102/10769986006002107)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang CR, Burns KH & Boeke JD 2012 Active transposition in genomes. Annual Review of Genetics 46 651675. (https://doi.org/10.1146/annurev-genet-110711-155616)

  • Khadivi F, Koruji M, Akbari M, Jabari A, Talebi A, Ashouri Movassagh S, Panahi Boroujeni A, Feizollahi N, Nikmahzar A & Pourahmadi M et al.2020 Application of platelet-rich plasma (PRP) improves self-renewal of human spermatogonial stem cells in two-dimensional and three-dimensional culture systems. Acta Histochemica 122 151627. (https://doi.org/10.1016/j.acthis.2020.151627)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kheil MH, Bahsoun R & Sharara FI 2022 Platelet-rich plasma: inconclusive evidence of reproductive outcomes in menopausal women. Journal of Assisted Reproduction and Genetics 39 19871991. (https://doi.org/10.1007/s10815-022-02554-5)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim MK, Yoon JA, Yoon SY, Park M, Lee WS, Lyu SW & Song H 2022 Human platelet-rich plasma facilitates angiogenesis to restore impaired uterine environments with Asherman's syndrome for embryo implantation and following pregnancy in mice. Cells 11 1549. (https://doi.org/10.3390/cells11091549)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Klugbauer N, Lacinova L, Flockerzi V & Hofmann F 1995 Structure and functional expression of a new member of the tetrodotoxin-sensitive voltage-activated sodium channel family from human neuroendocrine cells. EMBO Journal 14 10841090. (https://doi.org/10.1002/j.1460-2075.1995.tb07091.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kordowitzki P, Kranc W, Bryl R, Kempisty B, Skowronska A & Skowronski MT 2020 The relevance of aquaporins for the physiology, pathology, and aging of the female reproductive system in mammals. Cells 9 E2570. (https://doi.org/10.3390/cells9122570)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kreiling JA, Jones BC, Wood JG, De Cecco M, Criscione SW & Neretti N et al.2017 Contribution of retrotransposable elements to aging. In Human Retrotransposons in Health and Disease. Cham, CH: Springer. Cristofari GEd., pp. 297321. (https://doi.org/10.1007/978-3-319-48344-3_13)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li X & Li XD 2021 Integrative chemical biology approaches to deciphering the histone code: a problem-driven journey. Accounts of Chemical Research 54 37343747. (https://doi.org/10.1021/acs.accounts.1c00463)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Z, Zhang M, Zheng J, Tian Y, Zhang H, Tan Y, Li Q, Zhang J & Huang X 2021 Human umbilical cord mesenchymal stem cell-derived exosomes improve ovarian function and proliferation of premature ovarian insufficiency by regulating the Hippo signaling pathway. Frontiers in Endocrinology (Lausanne) 12 711902. (https://doi.org/10.3389/fendo.2021.711902)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu S, Strauss S, Adibi M, Mosca G, Yoshida S, Dello Ioio R, Runions A, Andersen TG, Grossmann G & Huijser P et al.2022 Cytokinin promotes growth cessation in the Arabidopsis root. Current Biology 32 19741985.e3. (https://doi.org/10.1016/j.cub.2022.03.019)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • López-Otín C, Blasco MA, Partridge L, Serrano M & Kroemer G 2013 The hallmarks of aging. Cell 153 11941217. (https://doi.org/10.1016/j.cell.2013.05.039)

  • Mai T, Markov GJ, Brady JJ, Palla A, Zeng H, Sebastiano V & Blau HM 2018 NKX3-1 is required for induced pluripotent stem cell reprogramming and can replace OCT4 in mouse and human iPSC induction. Nature Cell Biology 20 900908. (https://doi.org/10.1038/s41556-018-0136-x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mao J, Zhang Q, Ye X, Liu K & Liu L 2014 Efficient induction of pluripotent stem cells from granulosa cells by Oct4 and Sox2. Stem Cells and Development 23 779789. (https://doi.org/10.1089/scd.2013.0325)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Miyoshi N, Ishii H, Nagano H, Haraguchi N, Dewi DL, Kano Y, Nishikawa S, Tanemura M, Mimori K & Tanaka F et al.2011 Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell 8 633638. (https://doi.org/10.1016/j.stem.2011.05.001)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Montserrat N, Nivet E, Sancho-Martinez I, Hishida T, Kumar S, Miquel L, Cortina C, Hishida Y, Xia Y & Esteban CR et al.2013 Reprogramming of human fibroblasts to pluripotency with lineage specifiers. Cell Stem Cell 13 341350. (https://doi.org/10.1016/j.stem.2013.06.019)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moody SC, Wakitani S, Young JC, Western PS & Loveland KL 2020 Evidence that activin A directly modulates early human male germline differentiation status. Reproduction 160 141154. (https://doi.org/10.1530/REP-20-0095)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nielsen JL, Bakula D & Scheibye-Knudsen M 2022 Clinical trials targeting aging. Frontiers in Aging 3 820215. (https://doi.org/10.3389/fragi.2022.820215)

  • Petralia RS, Mattson MP & Yao PJ 2014 Aging and longevity in the simplest animals and the quest for immortality. Ageing Research Reviews 16 6682. (https://doi.org/10.1016/j.arr.2014.05.003)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pezzulo G, LaPalme J, Durant F & Levin M 2021 Bistability of somatic pattern memories: stochastic outcomes in bioelectric circuits underlying regeneration. Philosophical Transactions of the Royal Society of London Series B 376(1821) 20190765. (https://doi.org/10.1098/rstb.2019.0765)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ptashne M 1992 A genetic switch: phage and higher organisms. Cambridge, MA: Blackwell Publishing Scientific & Cell Press, pp. 1192. Available at: http://hdl.handle.net/10822/545304

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rajkovic A, Pangas SA, Ballow D, Suzumori N & Matzuk MM 2004 NOBOX deficiency disrupts early folliculogenesis and oocyte-specific gene expression. Science 305 11571159. (https://doi.org/10.1126/science.1099755)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rickers NS & Sills ES 2022 Is autologous platelet activation the key step in ovarian therapy for fertility recovery and menopause reversal? Taipei: Biomedicine.

  • Sassoli C, Garella R, Chellini F, Tani A, Pavan P, Bambi F, Zecchi-Orlandini S & Squecco R 2022 Platelet-rich plasma affects gap junctional features in myofibroblasts in vitro via vascular endothelial growth factor (VEGF)-A/VEGF receptor. Experimental Physiology 107 106121. (https://doi.org/10.1113/EP090052)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Saumell-Esnaola M, Delgado D, García Del Caño G, Beitia M, Sallés J, González-Burguera I, Sánchez P, López de Jesús M, Barrondo S & Sánchez M 2022 Isolation of platelet-derived exosomes from human platelet-rich plasma: biochemical and morphological characterization. International Journal of Molecular Sciences 23 2861. (https://doi.org/10.3390/ijms23052861)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schächner C, Merkl PE, Pilsl M, Schwank K, Hergert K, Kruse S, Milkereit P, Tschochner H & Griesenbeck J 2022 Establishment and maintenance of open ribosomal RNA gene chromatin states in eukaryotes. Methods in Molecular Biology 2533 2538. (https://doi.org/10.1007/978-1-0716-2501-9_2)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sendžikaitė G & Kelsey G 2019 The role and mechanisms of DNA methylation in the oocyte. Essays in Biochemistry 63 691705. (https://doi.org/10.1042/EBC20190043)

  • Shin JW, Lee E, Han S, Choe SA & Jeon OH 2022 Plasma proteomic signature of cellular senescence and markers of biological aging among postmenopausal women. Rejuvenation Research 25 141148. (https://doi.org/10.1089/rej.2022.0024)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES, Li X, Rickers NS, Wood SH & Palermo GD 2019 Metabolic and neurobehavioral response following intraovarian administration of autologous activated platelet-rich plasma: first qualitative data. Neuro Endocrinology Letters 39 427433. Available at: https://www.nel.edu/userfiles/articlesnew/1553033179_39_6_sills_427-pdf.pdf

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES, Rickers NS, Li X & Palermo GD 2018 First data on in vitro fertilization and blastocyst formation after intraovarian injection of calcium gluconate-activated autologous platelet rich plasma. Gynecological Endocrinology 34 756760. (https://doi.org/10.1080/09513590.2018.1445219)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES, Rickers NS & Wood SH 2020 Intraovarian insertion of autologous platelet growth factors as cell-free concentrate: Fertility recovery and first unassisted conception with term delivery at age over 40. International Journal of Reproductive Biomedicine 18 10811086. (https://doi.org/10.18502/ijrm.v18i12.8030)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES & Wood SH 2019 Autologous activated platelet-rich plasma injection into adult human ovary tissue: molecular mechanism, analysis, and discussion of reproductive response. Bioscience Reports 39 BSR20190805. (https://doi.org/10.1042/BSR20190805)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES & Wood SH 2021 Progress in human ovarian rejuvenation: Current platelet-rich plasma and condensed cytokine research activity by scope and international origin. Clinical and Experimental Reproductive Medicine 48 311315. (https://doi.org/10.5653/cerm.2021.04651)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sills ES & Wood SH 2022 Growth factors, gene activation, and cell recruitment: from intraovarian condensed platelet cytokines to denovo oocyte development. Journal of Clinical and Translational Research 8 4953. Available at: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8848765/

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Smith ZD & Meissner A 2013 DNA methylation: roles in mammalian development. Nature Reviews. Genetics 14 204220. (https://doi.org/10.1038/nrg3354)

  • Sommer RJ 2020 Phenotypic plasticity: From theory and genetics to current and future challenges. Genetics 215 113. (https://doi.org/10.1534/genetics.120.303163)

  • Sturm Á, Perczel A, Ivics Z & Vellai T 2017 The piwi-piRNA pathway: Road to immortality. Aging Cell 16 906911. (https://doi.org/10.1111/acel.12630)

  • Sturm Á & Vellai T 2022 How does maternal age affect genomic stability in the offspring? Aging Cell 21 e13612. (https://doi.org/10.1111/acel.13612)

  • Takahashi K & Yamanaka S 2006 Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126 663676. (https://doi.org/10.1016/j.cell.2006.07.024)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tremellen K & Pacella-Ince L 2022 An audit of clinical outcomes following ovarian administration of platelet-rich plasma (PRP) in women with severe diminished ovarian reserve. Australian and New Zealand Journal of Obstetrics and Gynaecology 62 767772. (https://doi.org/10.1111/ajo.13578)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Weng HP, Cheng YY, Lee HL, Hsu TY, Chang YT & Shen YA 2021 Enhanced platelet-rich plasma (ePRP) stimulates wound healing through effects on metabolic reprogramming in fibroblasts. International Journal of Molecular Sciences 22 12623. (https://doi.org/10.3390/ijms222312623)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xu Y, Wang X, Liu W & Lu W 2021 Thrombin-activated platelet-rich plasma enhances osteogenic differentiation of human periodontal ligament stem cells by activating SIRT1-mediated autophagy. European Journal of Medical Research 26 105. (https://doi.org/10.1186/s40001-021-00575-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yasmin R, Siraj S, Hassan A, Khan AR, Abbasi R & Ahmad N 2015 Epigenetic regulation of inflammatory cytokines and associated genes in human malignancies. Mediators of Inflammation 2015 201703. (https://doi.org/10.1155/2015/201703)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yin F, Yao J, Sancheti H, Feng T, Melcangi RC, Morgan TE, Finch CE, Pike CJ, Mack WJ & Cadenas E et al.2015 The perimenopausal aging transition in the female rat brain: decline in bioenergetic systems and synaptic plasticity. Neurobiology of Aging 36 22822295. (https://doi.org/10.1016/j.neurobiolaging.2015.03.013)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yoder JA, Walsh CP & Bestor TH 1997 Cytosine methylation and the ecology of intragenomic parasites. Trends in Genetics 13 335340. (https://doi.org/10.1016/s0168-9525(9701181-5)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang J, Fang L, Lu Z, Xiong J, Wu M, Shi L, Luo A & Wang S 2016 Are sirtuins markers of ovarian aging? Gene 575 680686. (https://doi.org/10.1016/j.gene.2015.09.043)

  • Zhang J, Zhang J, Zhang N, Li T, Zhou X, Jia J, Liang Y, Sun X & Chen H 2020 The effects of platelet-rich and platelet-poor plasma on biological characteristics of BM-MSCs in vitro. Analytical Cellular Pathology 2020 8546231. (https://doi.org/10.1155/2020/8546231)

    • PubMed
    • Search Google Scholar
    • Export Citation