Skip to main content
Cell Research logoLink to Cell Research
. 2023 Jan 2;33(1):11–29. doi: 10.1038/s41422-022-00718-7

Unraveling female reproductive senescence to enhance healthy longevity

Lu Dong 1,2,#, Daniel Boon Loong Teh 3,4,#, Brian Keith Kennedy 1,2,3,4,5,, Zhongwei Huang 4,5,6,7,
PMCID: PMC9810745  PMID: 36588114

Abstract

In a society where women often want successful careers and equal opportunities to men, the early nature of ovarian aging often forces women to make difficult life choices between career and family development. Fertility in women begins to decline after the age of 37 years and it is rare for pregnancies to occur after 45. This reproductive decline in women is inevitable and culminates in menopause, which is a major driver of age-related diseases. In a world where biomedical advances are leading to modifiable biological outcomes, it is time to focus on mitigating female reproductive senescence to maintain fertility and preserve age-related hormonal functions, with the goal of providing increased life choices and enhancing healthspan. To date, reproductive longevity research remains an understudied field. More needs to be done to unravel the biology of the ovarian follicles, which are the functional units of reproductive lifespan and are comprised of cell types including the oocyte (female gamete) and a group of specialized supporting somatic cells. Biological attempts to maintain the quality and quantity of follicles in animal models through manipulating pathways involved in aging can potentially prolong female reproductive lifespan and healthspan. Here, we summarize the molecular events driving ovarian aging and menopause and the interventional strategies to offset these events. Developing solutions to female reproductive senescence will open doors to discover ways to enhance true healthy longevity for both men and women.

Subject terms: Cell biology, Molecular biology

Introduction

In women, the inevitability of reproductive decline is accepted as a natural phenomenon, with consequences on the life choices that are made. Fertility begins to significantly decline after approximately the age of 37 years old (although considerable variation exists) and it is rare for pregnancy to occur after 45 years old. Moreover, menopause is a driver of age-related diseases. In a society where women often want to (and deserve to) have successful careers and equal opportunities to men, prevailing biology often forces choices between sub-optimal outcomes. In a world where biomedical advances are increasingly leading to modifiable biological outcomes, it is time to focus on female reproductive decline as a modifiable event, where maintenance of fertility and preservation of age-related hormonal functions are emphasized with the goal of providing increased life choices and healthspan.

Notably, women live longer than men in most parts of the world — an average of 4–7 years longer in developed countries.1,2 Yet, the female gonad, the ovary, ages exceptionally early and rapidly before any other parts of the female body system.36 Each woman is born with about 1–2 million oocytes in the form of primordial follicles in her ovaries. After birth, this pool of follicles declines gradually and continuously. This depletion process is accelerated and coupled with a decrease in oocyte quality after about 31 years of age,7 leading to a gradual loss of fecundity. At the same time, the production of the main ovarian gonadal steroid family, estrogens, declines with the depletion of ovarian follicles. Ultimately, when the number of ovarian follicles drops below 1000,7 the woman reaches natural sterility, also known as menopause, which occurs at an approximate age of 50 years.36,8,9

With increased longevity, women spend on average, nearly 40% of their lives in menopause, which is a clinical phenomenon whereby the number of ovarian follicles is so low that there are insufficient levels of estrogens to bring about further ovarian activity and stimulation of the womb lining to result in cyclical menstrual bleeding. This results in the cessation of periods. If a woman aged over 45 years has no spontaneous periods consecutively (and not pregnant) for more than 12 months, she is deemed to have reached clinical menopause. Unfortunately, 1% of all women suffer from a condition known as premature ovarian insufficiency (POI),10 a state whereby the end of reproductive lifespan occurs before they are 40 years old due to a premature and irreversible loss of ovarian follicles. Importantly, several age-related chronic diseases are disproportionately affected by the onset of menopause.11,12 For instance, there are heightened risks of neurocognitive decline,13 cardiovascular diseases (CVDs),14 metabolic dysfunction,15 sarcopenia,16 insulin resistance, osteoporosis11 and sexual dysfunction. In addition to the permanent loss of fertility, women are at increased susceptibility to cardio-metabolic diseases, leading to premature mortality.14,17

The ovarian follicle as the basic functional unit of reproductive lifespan and healthspan

Mammalian ovaries, with the ovarian follicles serving as the functional units, work in sync with the mature hypothalamic-pituitary system, known as the hypothalamic-pituitary-ovarian axis, regulate menstrual cycles and govern the reproductive lifespan and healthspan of a woman (Fig. 1). During the menstrual cycle, the ovarian follicles support the maturation of oocytes, which are the female gametes carrying all genetic information and storing nutrients essential for embryo development upon successful fertilization. An enhanced ovarian follicle pool and better follicular quality thus indicate a longer reproductive lifespan.18

Fig. 1. Ovarian follicles: the functional units of ovary for reproductive lifespan.

Fig. 1

Along the hypothalamic-pituitary-ovarian axis, the hypothalamus secretes gonadotropin-releasing hormone (GnRH) which travels down to stimulate the pituitary gland which in turn secretes follicle stimulating hormone (FSH) and luteinizing hormone (LH). FSH and LH reach the ovaries in the bloodstream to signal the development of ovarian follicles to produce estrogen and progesterone during the follicular and luteal phase of the menstrual cycle. Estrogen rises steadily via a positive feedback loop to result in LH surge from the anterior pituitary gland, leading to ovulation. In a “young ovary”, in each cycle, several resting primordial follicles with immature ova are activated. They develop during folliculogenesis and normally only one ovarian follicle will be “selected” as the dominant follicle and eventually releases the mature ovum (ovulation). The ruptured follicle then transforms into the corpus luteum and degenerates to form the corpus albicans if no implantation occurred. Follicular supporting cells such as the granulosa and theca cells, and the corpus luteum provide endocrine support necessary for ovulation, preparation for implantation and pregnancy, with the release of estrogen and progesterone (symbolized as blue dots). As a woman ages, the finite pool of primordial follicles depletes during each ovulatory cycle and along with constant follicular atresia, results in the degeneration and loss of ovarian follicles and their oocytes, becoming the “aged ovary”. The “aged ovary” shrinks due to age-related fibrosis and releases little estrogen and progesterone due to the extremely low number of viable ovarian follicles.

Ovarian follicles are comprised of somatic cells, known as the granulosa and thecal cells, surround the oocytes and support the growth and development of oocytes. They are the energy hub to produce essential reproductive hormones, progesterone and androgens produced by theca cells and estrogens produced by granulosa cells (Fig. 1).19 These reproductive hormones, especially estrogens, maintain female reproductive health and provide systemic beneficial effects on other biologic systems. Estrogen is the paramount hormone in regulating the female reproductive system and maintaining women’s health, with roles in the cardiovascular system, cognitive function, skin homeostasis and bone metabolism, as reviewed previously.2023 Similarly, progesterone, a hormone that is released by the corpus luteum following ovulation and plays a role in maintaining pregnancy, exerts its effects on the reproductive system, but also supports neuroregeneration and neuroprotection.24

The number of ovarian follicles is finite at birth in a baby girl, comprising the entire ovarian reserve for her lifetime.25 As the woman ages, the follicles are constantly recruited for maturation: they either reach ovulation (~400) or (the majority) undergo atresia.26 Along with this decline in follicle number, follicle quality also decreases with age. This has been proposed to be caused by abnormalities in meiotic spindle assembly and chromosomal distribution in oocytes, as well as mitochondrial dysfunction in both oocytes and the surrounding follicular cells. These biological changes are associated with advancing maternal age and accompanied by declining ovarian steroidogenesis functions due to dysfunctional ovarian folliculogenesis.2733 Notably, even in young women with diminished ovarian reserve, the chances of achieving high-quality embryos and successful pregnancy in in vitro fertilization (IVF) have been shown to be much greater compared to older women, despite obtaining similar numbers of eggs.34 Thus, both quality and quantity of ovarian follicles are important, and their irrevocable decline will result in the end of a woman’s reproductive lifespan and, ultimately, a decline in overall health. Therefore, if a woman is born with a very low number of ovarian follicles, or experiences faster depletion of healthy follicles, she will reach menopause earlier. This contrasts with men, who continue to produce spermatozoa (for fertility) and androgens in their testes throughout their lives, albeit with lower quantity and quality as they age.

Menopause: an evolutionary vestigial inheritance?

Menopause appears to decouple reproductive from somatic lifespan in women, leading to reproductive senescence primarily due to a sudden deprivation of serum estrogens.35 Furthermore, menopause is extremely rare and only known to be present in 2–3 species of mammals; humans being (Homo sapiens) is the only terrestrial mammal with menopause (Fig. 2).36 Interestingly, these species are associated with a longer lifespan as compared to most without a long post-reproductive lifespan.

Fig. 2. Post-reproductive lifespan across species.36.

Fig. 2

The human female is one of a few selected species of mammals with a significant post-reproductive lifespan. The significance of menopause is extensively being debated on whether it is a vestigial evolutionary inheritance or has crucial implication in conferring evolutionary survival of the fittest to our early days’ ancestors as been reproduced from Ellis et al.36 (Open access license: https://creativecommons.org/licenses/by/4.0/. Image legend has been shifted to the top left of the image from the original image).

Beyond the permanent loss of fertility, the hypoestrogenic environment due to menopause is a profound accelerator of aging in women. Importantly, the age of menopause is potentially hereditary, with genome-wide association studies demonstrating the presence of genetic variants linked to menopause that are involved in DNA repair and maintenance. One genetic variant is also linked to systemic aging.10 Interestingly, the human ovary, is thought to be the first organ to decline in function with age. The evolutionary history of menopause is thought to be attributed to the “Grandmother Hypothesis”,37,38 whereby energy expended for reproduction is ceased and redirected to tending to their young and future generations to sustain survival of the larger group, as studied in the menopausal female killer whale (Orcinus orca).39 As modern humans have gained longevity due to the shift from prehistoric ancestor roles of hunters-gatherers to current times of technological advancement and enhanced lifespan, menopause may be seen as a vestigial inheritance from our ancestors. Menopause itself, was only coined in the 1820’s, by French physician Charles-Pierre-Louis de Gardanne.40 At that time, the average female lifespan was around 40–50 years of age, which is congruent to what is currently known as age of menopause in women which is ~49–50 years of age.41

Until relatively recent times, unhygienic sanitation and lifestyle coupled with the lack of access of quality healthcare, including treatment of infectious diseases, and a range of other factors contributed to early mortality in a non-gender selective manner.4245 The relative differences between reproductive lifespan and overall lifespan were likely to be small on an evolutionary timeframe during that period; however, with societal and medical improvements these may have result in large differences between reproductive lifespan and healthspan in women from an evolutionary perspective.

There are selected mammals that have extended reproductive lifespan46 such as the fin whale (Balaenoptera physalus) and elephant (Loxodonta) (Fig. 2), with longevity closely linked to the homogametic sex, as compared to the heterogametic sex. Interestingly, however, as compared to humans’ closest relatives such as chimpanzees (Pan troglodytes),47 it seems that the duration of reproductive lifespan is somewhat conserved. Yet the lifespan of chimpanzee regardless in the wild or captivity is far shorter.48 In contrast, prolonged human (Homo sapiens) reproductive age is linked to longevity.49 The conservation of energy due to lower resting metabolic rate during menopause,50 may have contributed to the longevity in humans, as compared to our closer vertebrate species, while this may not necessarily have translated to healthspan in women with increasing health problems associated with earlier menopause and after the onset of menopause.41

Pregnancies in women of advanced maternal age are linked to increased risks of obstetrical complications in the mother, such as gestational diabetes, pre-eclampsia, hypertension, and increased risks of fetal problems such as aneuploidy, e.g., Down’s Syndrome and other congenital malformations.5155 This is associated with the decline in ovarian follicle quality, which results in oocytes of poorer quality during maternal aging. Therefore, reproductive longevity research would have to address both the extension of reproductive lifespan and the improvement of reproductive healthspan, such that women of advanced maternal age can achieve healthy pregnancies and healthy babies.

Reproductive senescence, hypoestrogenism and inflammation as the driver of aging

Indeed, humans are just a handful of species with an evolutionary divergence of somatic senescence from reproductive senescence, with reaching menopause as the significant driver of aging in women. As discussed above, the deprivation of estrogens from the ovarian follicles dictates this reproductive inevitability. Estrogen confers cardio-protective effects, through improving circulation and vascular health, maintaining oxidative balance, while reducing fibrosis and arterial stiffness in the female vasculature (Fig. 3).5661 In preclinical models, estrogen acts as an anti-inflammatory agent, retarding CVD onset and progression, and this appears to be both sexually dimorphic and age sensitive, which explains the lower risk of CVD in premenopausal women compared to men and postmenopausal women.62 Epidemiological evidence correlating chronic diseases to the onset of menopause suggests that women in the reproductive age group are less prone to these comorbidities when compared to men.6366 Consequently, with the postmenopausal decline in estrogen levels, women become predisposed to chronic inflammatory conditions such as atherosclerosis and type 2 diabetes.6469 Estrogen directly binds to the estrogen receptors (ERs), ERα or ERβ,70 leading to their translocation into the nucleus to target genes driven by promoters containing estrogen responsive elements (EREs) and disrupting the inflammatory cascade by preventing NF-κB transcriptional activation.71 Disruption of NF-κB has a profound effect on the synthesis and secretion of pro-inflammatory cytokines such as TNFα, IL6, and IL1β. Furthermore, estrogen (exogenous in vivo replacement of 17β-Estradiol, but not progesterone) modulates the cell surface expression of Toll-like receptor 4 (TLR4) in macrophages, which is critical to the pro-inflammatory polarization (M1) phenotype and accompanying cytokine secretion.72,73

Fig. 3. The post-reproductive lifespan is a result of a hypoestrogenic environment that has pleiotropic health effects.

Fig. 3

However, the biological significance of the post-reproductive lifespan in women remains unknown.

Reproductive aging and inflammation on musculoskeletal health

Expression of ERs occurs in nearly all musculoskeletal and connective tissues, such as ligament and tendons.74,75 As the expression of ERs is ubiquitous in satellite cells (adult muscle stem cells) of muscle fibers, estrogen stimulates the activation and proliferation of these quiescent stem cells (satellite cells) during muscle repair or injury.76 Although both ERα and ERβ are present, ERα contributes to the critical signaling role of muscle stem cell maintenance and muscle regeneration in women.77 Systematically, chronic inflammation also affects the rejuvenation capability of satellite cells.78 In contrast, while estrogen is able to improve muscle mass and function, female athletes are known to experience significantly higher incidences of anterior cruciate ligament ruptures (due to increased laxity) during the different phases of the menstrual cycle due to the high estrogen levels, when compared to their male counterparts,78 whereas, menopause is linked to an elevated risk of musculoskeletal trauma and sarcopenia. This implies that, while estrogen is beneficial directly towards muscle health, it could reduce the stiffness of connective tissues, thereby reducing function and promoting injury.74

Bone mass integrity is maintained through the balance between bone resorption by osteoclasts and bone formation by osteoblasts.79 Osteoporosis is preceded by the micro-architectural loss of bone mass, which gradually reduces the ability of the bone to support body weight. Postmenopausal women are at higher risk of developing osteoporosis, and they form the largest number of osteoporotic cases.80,81 Serum estrogen levels are directly related to bone mineral density and inversely linked to bone fracture risks.82 Fundamentally, a hypoestrogenic environment tilts the balance of bone towards resorption, as estrogen exerts profound effects on the development and survivability of both osteoblasts and osteoclasts.83 Estrogen stimulates the maturation of osteoblasts through upregulation of the TGFβ signaling pathway, which is an apoptotic inducer through Fas ligand in osteoclasts.79,80,84,85 Furthermore, ERs are highly expressed in osteoblastic lineage cells, with ERα being the more prominent receptor. In ERα-knockout female mice, significant bone loss was observed, a phenotype similar to ovariectomized rodents.8689 ERα also confers selective suppression of the receptor activator of NF-κβ (RANKL), a critical cytokine in bone resorption that is secreted by hematopoietic and mesenchymal lineages.88

In estrogen deficient states, RANKL upregulation is seen in B lymphocytes.90 Conditional suppression of RANKL in B lymphocytes but not T lymphocytes, ablated the signs of bone loss in mice.90 Nonetheless, ERα expression is also found in T lymphocytes and RANKL is secreted from T lymphocytes as well. Although deletion of RANKL from T lymphocytes did little to protect from bone loss, estrogens enhance T cell activation and proliferation.91 T lymphocytes in return stimulate osteoclast differentiation and bone resorption, while secreting inflammatory cytokines such as TNFα and IL6, which further exacerbate bone loss.9193 T lymphocytes therefore could elicit a RANKL-independent, yet synergistic, pathway in osteolysis through the secretion of TNFα.92 In fact, TNFα is equipotent to RANKL in mediating osteoclastogenesis, further exerting synergistic effects in the presence of RANKL.92 Collectively, evidence from preclinical studies suggest that estrogen deficiency-induced postmenopausal osteoporosis has a strong immunomodulatory component intertwined with sex steroid actions. Therefore, an immunomodulatory effect from the precursors of T and B cells is likely to be implicated in postmenopausal osteoporosis with RANKL being suggested to be the major contributory factor to hypoestrogenism-induced osteoporosis in bone lining cells.88

Calcium makes up the major component in bone that provides strength and structure,94 and the absorption of calcium from the gut is very much reliant on the availability of vitamin D in the body.95 Aging is related to an increase in the production of vitamin D96 but malabsorption of calcium, which has a compounding effect on the onset of osteoporosis.97 Postmenopausal women suffer a negative calcium balance that slowly stabilizes over the years, but remains tilted towards calcium loss.98 As natural aging reduces the efficacy of vitamin D production in the liver and skin, it further aggravates the onset of androgenic-induced osteoporosis in women. Therefore, dietary supplementation of vitamin D and calcium, not either or, is only a temporary solution for postmenopausal women at high risk of osteoporosis. Similarly, data from Women’s Health Initiative (WHI) studies demonstrate that menopausal hormone replacement therapy (HRT)99,100 causes a significant reduction in the incidence of bone fractures in postmenopausal women.99,101 However, the use of menopausal HRT, including transient and lowest dose usage, in mediating benefits on musculoskeletal health needs to be weighed with increased risks of breast cancer and stroke. Several reviews have discussed this issue in more detail.100,102

Reproductive aging and inflammation on cardio-metabolic health

Menopause presents a void in sex hormone-mediated homeostasis of glucose and lipid metabolism, which has profound implications in obesity and glucose impairment. Sex hormones are known to regulate the distribution of visceral fat in humans.103105 Fat distribution is altered in pre- and postmenopausal women, where subcutaneous fat is progressively being displaced by abdominal fat. Interestingly, adipocytes express both ERα and ERβ, yet only ERα is found to be in brown adipocytes.106,107 Brown adipose tissue maintains energy expenditure by enhancing lipid and glucose metabolism.108 It is also part of the endocrine system that secretes adipokines, which regulate inflammatory responses and confer cardio-protection.109,110 Hence, alteration of ERα in animal models leads to central obesity and diabetes, and in humans this change in fat deposition is also associated with the development of CVD risk factors.111,112

During menopause, changes in metabolism include lower energy expenditure, predisposition to central adiposity and an impairment of insulin sensitivity. Clinical evidence is mixed on whether HRT confers CVD protection in postmenopausal women. The Nurse’s Health Study (NHS), a prospective longitudinal study spanning 20 years recruited 121,000 female nurses, with 70,533 being postmenopausal, suggested that HRT significantly reduced the risks of CVD, as compared to non-users.105 In contrast, the Heart and Estrogen/Progesterone Replacement Study (HERS), a randomized controlled trial demonstrated that administering conjugated equine estrogens (CEE) and medroxyprogesterone acetate confers no beneficial effects.102 Likewise, two subsequent randomized controlled trials of the WHI involving either estrogen + progesterone (WHI E + P) (16,608 postmenopausal women were assessed)100 or estrogen alone (WHI E)99 (10,739 postmenopausal women who had undergone hysterectomy were followed) were stopped abruptly following an elevated risk of coronary heart disease in the former trial and an increase in incidence of stroke in the latter. Successive smaller studies, the Danish Osteoporosis Prevention Study (DOPS)113 and Early versus Late Intervention Trial with Estradiol (ELITE),114 were more promising in demonstrating the protective effects of menopausal hormone therapy in reversing the risks of CVD and atherosclerosis. DOPS followed the study population of postmenopausal women for 10 years, and showed that estrogen treatment given to women, significantly suppressed their risks of mortality, CVD, and heart failure. Notably, in DOPS there was no significant elevation of cancer risks.

The optimal period of estrogen rescue is likely to be at the early postmenopausal stage, as demonstrated in ELITE, where there were reduced atherosclerotic events noted at early, but not late stage of postmenopausal intervention. In the Kronos Early Estrogen Prevention Study (KEEPS),115 only relief of vasomotor symptoms was observed, with a beneficial trend in CVD. In DOPS, ELITE and KEEPS, no obvious adverse effects were observed in the use of estrogens. Findings in ELITE reinforced the “Timing Hypothesis”, in which estrogen’s Jekyll and Hyde role is dependent on intervention prior to the onset of erosion of coronary plaque, which can be worsened by hormonal therapy.116 Impairment in glucose metabolism represents a more significant phenotype of menopause. Despite the poor outcome of the HERS and WHI trials in de-escalating the onset of CVD risks, these studies highlighted the significant role that estrogens have in reducing the onset of type-2 diabetes,99,100,102 a significant risk factor for CVD. Additionally, women who underwent estrogen replacement therapy were observed to have lower hemoglobin A1c (HbA1c), a risk factor for diabetes.117,118

Interventions with HRT are directly beneficial to women who suffer from premature POI, while the beneficial effects in women who attain menopause at the expected age of about 50 years likely depend on treatment inception soon after menopause. Potential health benefits by these hormones can be maintained, such as protective effects against CVDs,119,120 brain aging121,122 and osteoporosis,123 thus enhancing the women’s healthspan, which is especially true for women who suffer from POI (i.e., menopause before they are 40 years old). It seems that in older women who were more than 5–10 years post menopause and above 60 years of age, HRT can be detrimental and cause increased risks of breast cancer and stroke with long-term use, as demonstrated in the WHI studies, leading to controversies about its use.124127 At the same time, several systematic reviews have failed to demonstrate protective effects of HRT against age-related health risks such as CVD events128 and cognitive decline129 in postmenopausal women, as HRT does not reverse processes that have already aged. However, it remains essential for women with premature and early ovarian insufficiency who require HRT for maintenance of their healthspan before the age of natural menopause (49–50 years).

The estimated life expectancy has been steadily improved to an average of 72.6 years (and 79.4 years in developed regions) for human beings.1 However, there has not been an equivalent improvement in healthspan, as the older individuals suffer from frailty and other aging-related medical conditions that affect multiple organs in the body.130132 This is further accentuated by ovarian or reproductive senescence. This partial uncoupling of healthspan from lifespan has raised the idea that for people beyond a certain age, i.e., 65 years old, life extension should not be the primary goal.133,134 Thus, a major need exists to define the critical period for intervention in women to enhance reproductive healthspan and to reduce all-cause morbidity. This likely involves mimicking the beneficial effects of estrogen postmenopausally, while avoiding the associated side effects. This will be one ultimate goal of reproductive longevity research, which will not only attempt to tackle the issue of reproductive lifespan, but also enhance female healthspan and lifespan.

Strategies to delay or even reverse ovarian aging as means to maintain reproductive lifespan and healthspan

Assisted reproductive technology (ART), established in the late 20th century, has become more popular and accessible now11 to circumvent infertility problems due to  the loss of oocyte quantity and/or quality to difficulty in fertilization and embryogenesis. Adjunct strategies, such as oocyte and ovarian tissue cryopreservation, have also matured, leading to rising success rates and utility.135,136 However, these strategies do have their limitations as they circumvent, instead of directly targeting, the root cause of fertility decline — ovarian aging. Thus, while allowing a woman to try to conceive, these strategies are unable to guarantee reproductive success as oocyte quality constantly decreases with age and advanced maternal age has become the most common factor for IVF failure.137,138 Furthermore, ART does not restore or maintain the levels and protective effects of ovarian gonadal steroids, such as estrogens.

Given that ovarian follicles are the functional units of ovaries, studies had attempted to enhance ovarian health and prevent reproductive aging through employment of various interventions studied in the context of aging and reported to enhance lifespan in animal models.139 These studies report molecules/compounds that target specific signaling pathways, spanning from antioxidants, precursors of oxidative stress pathways, mitochondrial chain precursors, isoflavones, polyphenol derivatives, and plant-based compounds, to commonly used medications like metformin and, recently, the mammalian target of rapamycin (mTOR) inhibitors (Table 1). Interestingly, most of this exploratory work was performed in rodent models, and results may differ between these models and humans. Rodent models provide key insights into possible human interventions, but follow-up human studies are needed. This is in part because female rodents and women share progressively increasing irregularity in ovulatory cycles (known as estrous cycle in rodents) and increasing fetal aneuploidy, as decline of oocyte quality and quantity becomes imminent with age, although female mice do not experience menopause like women.140 Primate models can be employed to study reproductive senescence, although they are expensive and time-consuming.

Table 1.

Pharmacological interventions for female reproductive longevity under research in mammalian models and clinical studies.

Pharmacological intervention Strain and ages/timings of outcome measurementsa (all animals used were female) Dosing regimen Outcomes at the oocyte levelb Outcomes at the ovarian follicle levelb Outcomes related to the estrous cyclesb Litter size and other mating outcomesb Other outcomesb Clinical evidence and other remarks

NAC155

NAC is a prodrug of L-cysteine, which is a precursor of the biologic antioxidant glutathione. NAC is thus used as an antioxidant by replenishing the glutathione store. NAC is approved for treating paracetamol overdose and has been widely researched for multiple conditions related to oxidative stress.

– Kunming mice,

– At 7–8 months and 12 months of age

Drinking water supplemented with 0.1 mM and 1 mM NAC from 4 weeks of age for 6–12 months

– Decrease in the number of poor-quality oocytes at 12 months of age

– No change in the number of ovulated oocytes

NA NA – Increase in litter size at 7–8 months of age – Increase in telomere length and telomerase activity in ovaries NA

Vitamin C232

Vitamin C, also known as L-ascorbic acid, is a water-soluble vitamin. Acting as an enzyme substrate and/or cofactor, it is required for the biosynthesis of collagen, L-carnitine, and certain neurotransmitters and involved in protein metabolism. It is also a physiological antioxidant by directly scavenging free radicals and indirectly restoring other antioxidants including α-tocopherol (vitamin E).

– NMRI mice

– After 8, 12 and 33 weeks of treatment

Oral gavage of 150 mg/kg vitamin C daily in young adults (25–30 g, age not specified) for 33 weeks – No change in the total volume of oocytes in antral follicles

– Increase in the number of granulosa cells

– Increase in the total number of primordial, primary, and antral follicles at 12 and 33 weeks of treatment

NA NA – Increase in ovarian volume NA

Vitamins C and E157

The vitamin E group, comprising of tocochromanols (tocopherols and tocotrienols), is the major hydrophobic vitamin exclusively obtained from diet. It reacts with unstable lipid radicals produced in lipid peroxidation to protect cell membranes from oxidative damage.

– F1 hybrid mice (C57Bl/6JIco female × CBA/JIco male)

– At the ages of 40–42, 50–52, and 57–62 weeks

Diet supplemented with 10 g/kg vitamin C and 0.6 g/kg vitamin E from first day of weaning or from 32 weeks of age

– Increase in oocyte number

– Improvement in oocyte quality by a decrease in age-related chromosomal distribution abnormalities in oocytes

NA – No effect except a decrease in the number of estrus cycles in the mice treated from weaning in comparison to the mice treated from 32 weeks of age NA NA NA

Genistein156,233

Genistein is a naturally occurring isoflavone derived from soy products. It exhibits multiple biological activities including anti-aging, antioxidant, phytoestrogenic and antineoplastic activities.

– Sprague-Dawley rats

– At 15 months of age156

Intragastric gavage of 160 mg/kg genistein daily from 11 months of age for 4 months NA

– Increase in the number of healthy follicles

– Decrease in the number of atretic follicles

– Increase in the number, but no change in the percentage of primordial follicles

– Decrease in the percentage of primary follicles

– Maintenance of estrous cycle NA NA In a study234 feeding adult female cynomolgus monkeys at 9–15 years of age saturated fat and cholesterol-containing diets, the diet with soy protein source containing genistein has led to higher numbers of primary and secondary follicles compared to the other diet with casein- lactalbumin source.

– Sprague-Dawley rats

– At 4 months and 15 months of age233

Intragastric gavage of 160 mg/kg genistein daily from 3 months of age for 4 weeks and 11 months of age for 4 months NA

– Increase in the number of healthy follicles

– Decrease in the number of atretic follicles

– Increase in the numbers of primordial and antral follicles at 15 months of age

– Maintenance of estrous cycle NA NA

Resveratrol235,156,167

Resveratrol is a natural polyphenol derived from red grapes and found in several other plants. It has been widely investigated as a therapeutic treatment for multiple diseases due to its antioxidant, anti-aging, anti-inflammatory, and anti-neoplastic properties. Resveratrol has been shown to scavenge ROS, enhance antioxidant biosynthesis and induce sirtuin-1 signaling.

– Sprague-Dawley rats

– At 15 months of age156

Intragastric gavage of 25 mg/kg resveratrol daily from 11 months of age for 4 months NA

– Increase in the number of healthy follicles

– Decrease in the number of atretic follicles

– Increase in the number, but no change in the percentage of primordial follicles

– Decrease in the percentage of primary follicles

– Maintenance of estrous cycle NA NA A cross-sectional retrospective study167 showed a strong association between resveratrol oral supplementation (200 mg/day) in women during IVF-embryo transfer and a decrease in clinical pregnancy rate.

– C57 mice

– At 14–16 months of age235

Drinking water supplemented with 30 mg/L resveratrol from 6 weeks of age for 6–12 months

– Increase in oocyte number

– Improvement in oocyte quality by better spindle morphology and chromosome alignment

– Increases in the numbers of primordial and primary follicles, as well as secondary and antral follicles

– No change in atretic follicle number

NA – Increase in litter size – Prevention of telomere shortening and improvement in telomerase activity in ovaries

CoQ10237

CoQ10, also known as ubidecarenone, is a naturally occurring component in cell membranes. It is an essential electron carrier in mitochondrial respiratory chain. CoQ10 also functions as an antioxidant via inhibition of lipid peroxidation as well as protein and DNA oxidation.

– ICR mice

– At 12 months of age

Subcutaneous injection of 22 mg/kg CoQ10, three times a week from 9 months of age for 12–13 weeks

– Improvement in oocyte quality by reduction in oocyte spindle defects

– Improvement in oocyte mitochondrial function

– Increase in the numbers of primordial, primary, and secondary follicles NA – Increase in litter size – Increase in ovulation rate In the same study,236 resveratrol (10 mg/kg) or lipoic acid (33 mg/kg) using the same dosing regimen and mouse strain did not lead to an increase in the ovulation rate.

Apocynin237,238

Apocynin, also known as acetovanillone, is a plant-derived polyphenol with multiple biological activities. It can suppress superoxide production by inhibiting NADPH oxidase.

– C57BL/6 J mice

– At 15–19 and 35–49 weeks of age237

Drinking water supplemented with 5 mM apocynin from 8–12 weeks or 38–42 weeks of age for 7 weeks NA – No change in the number or percentage of follicles at different stages NA NA

– Improvement in redox homeostasis and reversal of age-related protein carbonylation increase in ovaries and uteri

– Normalization of age-related inflammation factors and collagen expression in ovaries

NA

– C57BL/6 J mice

– At 8–12 weeks and 38–42 weeks of age238

Drinking water supplemented with 5 mM apocynin from 6 weeks of age until 8–12 weeks or 38–42 weeks of age NA NA NA

– Increase in litter size

– Increase in the number of uterine implantation sites

– Improvement in redox homeostasis and reversal of age-related protein carbonylation increase in uterus

Acetyl carnitine and lipoic acid239

Both acetyl carnitine and lipoic acid are natural compounds in the body and have demonstrated anti-inflammatory and antioxidative properties. Acetyl carnitine is formed by the addition of an acetyl group to the amino acid carnitine while lipoic acid is derived from octanoic acid. Acetyl carnitine facilitates fatty acid metabolism while lipoic acid has been shown to increase glutathione synthesis and regenerate vitamins C and E.

– C57BL/6 mice

– After 3, 6, 9 and 12 weeks of treatment

Drinking water supplemented with 100 mg/L acetyl carnitine and 40 mg/L lipoic acid from 3 weeks of age for 3, 6, 9 or 12 months of treatment

– Increase in oocyte number

– Improvement in oocyte quality by reduction in oocyte chromosomal and spindle abnormalities

– Improvement in oocyte mitochondrial function

NA NA NA NA NA

Dimethylfumarate (DMF)240

DMF is a lipophilic drug approved for treating psoriasis and sclerosis. It has been shown to activate the Nrf2 pathway and possess immunomodulatory and antioxidant properties.

– BALB/c mice

– At 48 weeks of age

Oral administration of 50 mg/kg DMF daily from 32 weeks of age for 16 weeks – Increase in oocyte number

– Increase in the number of primordial follicles

– No change in the numbers of primary, secondary, and antral follicles

NA NA

– Elevation of serum AMH

– Increase in antioxidant levels and decrease in oxidative stress in ovaries

– Increase in telomere mRNA and protein levels in ovaries

NA

Catalpol241

Catalpol, a plant-derived iridoid glucoside, has been shown to have antioxidative, anti-inflammatory, anti-apoptosis, and neuroprotective properties.

– Sprague-Dawley rats

– At 15 months of age

Oral gavage of 1, 3 or 5 mg/kg catalpol daily from 14 months of age for 4 weeks NA NA NA NA

– Alleviation in ovarian weight loss and structural abnormalities

– Rejuvenation of ovarian granulocytes

– Increase in serum estradiol and progesterone levels but decrease in serum follicle-stimulating and luteinizing hormone levels

NA

Metformin177,179,242

Metformin is a first-line biguanide antidiabetic drug. It has been shown to reduce hepatic glucose production and intestinal glucose absorption and improve insulin sensitivity. Metformin is also used to treat polycystic ovarian syndrome and investigated as a longevity drug. Proposed mechanisms of its life-extending effects include activation of the AMPK pathway, as well as modulation of the gut microbiota and DNA methylation.

– C57BL/6

– At 54 weeks of age177

Diet supplemented with 100 mg/kg metformin from 28 weeks of age for half a year NA – Increase in the numbers of primordial and primary follicles – Maintenance of estrous cycle NA

– Reduction in ovarian oxidative damage and senescence marker P16

– Maintenance of serum estradiol level

A prospective randomized trial242 found that female IVF repeaters without PCOS and with a mean age of about 39 years who took 500 mg/day metformin for 8–12 weeks before and during ovarian stimulation have higher ongoing pregnancy and implantation rates compared to those untreated.

– Wistar albino rats

– At 12 weeks of age179

Oral gavage of 100 and 200 mg/kg/day metformin from 8 weeks of age for four weeks NA – No significant change in the number of primordial, primary, or total follicles, except an increase in secondary follicle count with 200 mg/kg/day dose NA NA – Decrease in endometrial thickness
2-DG180 2-DG is a glucose derivative that competitively inhibits glycolysis, thus mimicking glucose restriction. It is also an investigational anticancer and antiviral drug.

– C57BL/6 mice

– At 7 weeks of age

Intraperitoneal injection of 100, 300 or 600 mg/kg 2-DG daily from 5 weeks of age for 2 weeks NA – Reduction in follicular activation by a decrease in the type 3a primary follicle count NA NA NA NA

NAD+ boosters186,187

NAD+ is naturally synthesized in the body. It is a central coenzyme in energy metabolism and an essential redox cofactor. The NAD+ level decreases with age and NAD+ repletion via the use of NAD+ precursors has been investigated as a therapeutic approach in multiple age-associated conditions, such as cardiovascular and neurodegenerative diseases

– Mainly C57BL/6 mice

– At 12–16 months of age187

Drinking water supplemented with 0.5 or 2 g/L of nicotinamide mononucleotide from 12 or 13 months of age for 4 weeks

– Improvement in oocyte quality by better oocyte spindle assembly

– Increase in oocyte yield, oocyte diameter and blastocyst formation rates (2 g/L)

NA NA

– Increase in litter size

– Increase in pregnancy rate and live birth rate (0.5 g/L)

NA NA

– C57BL/6 mice

– At 12 months of age186

Drinking water supplemented with 400 mg/kg/day of nicotinamide riboside from 8 months of age for 4 months

– Improvement in oocyte quality by better oocyte spindle assembly

– Improvement in mitochondrial functions in ovaries and oocytes

– Increase in the numbers of primordial, primary, and total follicles – Maintenance of estrous cycle – Increase in litter size – Increase in ovary size and the number of ovulated cumulus–oocyte complexes after superovulation

α-ketoglutarate (AKG)243

AKG is a key intermediate in the Krebs cycle, a nitrogen scavenger and a precursor of glutamate and glutamine. AKG can enhance protein synthesis, bone development and immune responses. It has also demonstrated life-extending effect by inhibiting ATP synthase and TOR.

– ICR mouse

– At 8 and 14 months of age

Drinking water supplemented with 2, 10, 25, and 50 mM (optimal: 10 mM) AKG from 2 to 14 months of age

– Increase in oocyte number

– Improvement in oocyte quality by reduction in abnormalities in mitochondrial distribution and spindle arrangement

– Increase in the number of primordial and primary follicles

– No significant change in the numbers of secondary, mature and atretic follicles

– No significant effect in estrous cycle

– Increase in litter size, pregnancy rate and pregnancy outcomes

– Decrease in neonatal death rate

– Reduction in telomere shortening NA

Melatonin218229

Melatonin is an amine neurohormone primarily secreted by pineal gland at night. Besides its role of maintaining circadian rhythm, melatonin also acts as a potent antioxidant and a direct free radical scavenger, speculatively through the Nrf2-ARE pathway and sirtuin activation. Moreover, melatonin has been shown to increase lifespan in some animal models.

– Holtzman rats

– At 75, 180 and 380 days of age218

Drinking water supplemented with 10 μg/mL melatonin from 10 days of age, either only at night or continuously for one year NA – No change in the number of primordial follicles – Decrease in estrous cycles with abnormal length with treatment only at night NA NA Several clinical trials224229 have investigated the effects of oral administration of melatonin, usually at the dose of 3 mg daily, in women with infertility or a history of IVF failures. Positive changes of gonadotropins, decrease in degenerate oocytes, improvement in oxidative balance and oocyte quality and/or a slight increase in IVF fertilization rate have been found in some studies,224226,229 However, other studies,226228 including one using a high dose of 8 mg twice daily, have failed to demonstrate an increase in clinical pregnancy rate, live birth rate or quality or quality of oocytes during IVF.

– Wistar rats

– At a) 5, b) 15 and c) 24 months of age219

Subcutaneous injection of 150 μg/100 g daily

from a) 3, b) 13 and c) 22 months of age for two months

– b) Increase in oocyte volume

– a) b) c) No change in oocyte density

NA – b) Improvement in estrous cycle regularity NA

– b) Increase in ovarian volume

– a) c) No change in ovarian volume

– a) b) Maintenance of serum estradiol level

– Kunming mice

– At 14–16 months of age221

Drinking water supplemented with 10 mg/kg melatonin from 2–3 months of age for 6–12 months

– Increase in oocyte number

– Improvement in oocyte quality by better spindle morphology and chromosome alignment

– Increase in primordial follicle count

– Decrease in atretic follicle count

– No change in growing or mature follicle counts

NA – Slight increase in litter size

– Improvement in ovarian mitochondrial antioxidant activity

– Increase in telomere length

– Suppression of ovarian apoptosis

– ICR mice

– At 43 and 53 weeks of age220

Drinking water supplemented with 100 μg/mL melatonin from 10 to 43 weeks of age – Increase in the number of ovulated oocytes – Increase in numbers of primordial, primary, and antral follicles NA NA

– Increase in telomere length

– Increase in rates of fertilization and blastocyst formation following IVF

– Kunming mice

– At 24–48 weeks of age222

Drinking water supplemented with 10−3, 10−5, 10−7 mol/L (optimal: 10−5 mol/L) melatonin from 8 to 24–48 weeks of age

– Increase in oocyte number

– Improvement in oocyte quality by improved morphology and blastocyst formation

– Reduction of ROS formation in oocytes

NA NA – Increase in litter size

– Improvement in ovarian antioxidant capacity

– Increase in the number of formed blastocysts and litter size following IVF

– Kunming mice

– On a) postnatal day 6 and 9 and b) postnatal day 15, 17, 19 and 21223

Injection of 1 and 15 mg/kg melatonin daily a) from postnatal day 3 to 9 and b) from postnatal day 10 to 21

– b) No change in oocyte diameter

– b) Decrease in oocyte number after superovulation

– a) Decrease in the number of activated follicles, but no change in atretic follicle number

– b) Decrease in the numbers of type 5b follicles and atretic follicles

– b) No change in estrous cycle – b) Decrease in the number of implanted embryos with bigger embryo size

– a) Improvement in ovarian antioxidant capacity

– b) Decrease in Fshr and Lhcgr gene expressions

Rapamycin4,6,207,208,210,244

Rapamycin, also known as sirolimus, is a natural anti-fungal macrolide produced by the bacterium Streptomyces hygroscropicus. Currently approved as an immunosuppressant, it has demonstrated therapeutic potential in other aspects, such as antiproliferation and immunomodulation in cancer treatment. Rapamycin has also been shown to extend both lifespan and healthspan in multiple animal models by inhibiting the target of rapamycin (TOR), a highly conserved key protein kinase regulating metabolism and physiology.

– C57BL/6 mice

– At 8 weeks of age244

Intraperitoneal injection of 2 doses of 5 and 50 mg/kg rapamycin from 8 weeks of age for two consecutive days – Reduction in ovulated eggs with unaffected quality – No change in follicle reserve NA NA NA NA

– Sprague-Dawley rats

– At 20 weeks of age207

Intraperitoneal injection of 5 mg/kg rapamycin every other day from 10 weeks of age for 10 weeks NA

– Suppression of primordial follicle activation by an increase in primordial follicle number

– Decrease in numbers of antral and atretic follicles and corpora lutea

– Disturbance of estrous cycle, during or just after treatment NA – Reduction in ovarian weight

– Adult rats

– Age not specified210

Intraperitoneal injection of 5 mg/kg rapamycin every other day for 10 weeks NA

– Suppression of primordial follicle activation by an increase in primordial follicle number

– Decrease in numbers of antral and atretic follicles and corpora lutea

– Disturbance of estrous cycle, during or just after treatment – Failure of being impregnated NA

– C57BL/6 mice

– At a) postnatal day 7 or b) postnatal day 21208

Intraperitoneal injection of 1 mg/kg rapamycin a) on postnatal day 2 and b) from postnatal day 7 and then every other day until postnatal day 21 NA – Suppression of primordial follicle activation by an increase in primordial and transitory follicle numbers and decrease in the numbers of primary and activated follicles NA NA NA

– CD1 mice

– Just after treatment, 2 months after treatment, and 16 months of age4

Intraperitoneal injection of 2 mg/kg rapamycin daily from 8 weeks or 8 months of age for 2 weeks

– No change in oocyte number

– Improvement in oocyte quality by morphology and spindle arrangement

– Suppression of primordial follicle activation by an increase in primordial follicle number and decrease in the numbers of primary, secondary, and antral follicles

– Disturbance of estrous cycle, during or just after treatment

– Re-normalization and maintenance of estrous cycle and hormone biogenesis 2 months post treatment

– Improvement in late-life fecundity NA

– C57BL/6 mice

– At 130 days of age6

Intraperitoneal injection of 4 mg/kg rapamycin every other day from 37 days of age for 93 days NA – Suppression of primordial follicle activation by an increase in primordial follicle number and decrease in the numbers of primary, secondary, and tertiary follicles NA NA NA

aIn vivo studies on aged wild-type animals and clinical studies treating infertile women without a particular confounding disease (e.g., PCOS, POI).

bOutcomes in comparison with untreated age-matched control in each study unless otherwise stated.

Utilizing agents studied in the context of general aging for correction of reproductive aging

Referring to Table 1, antioxidants represent the biggest category of compounds studied for reproductive aging, as the free radical theory has been the classical aging theory that attributes aging phenomena to accumulated cellular oxidative stress.141 According to this theory, the accumulation of reactive oxygen species (ROS) leads to ovarian oxidative stress and changes in ovarian microenvironment, resulting in cellular senescence and a decrease in oocyte quality and quantity.142145 Indeed, oxidative stress has been shown to be associated with aging-related oocyte deterioration.146148 In addition, previous studies found an increase in ROS and a decrease in levels of antioxidants in oocytes of older women receiving IVF, which are also associated with ART failure.149151 Antioxidants such as N-acetyl-L-cysteine (NAC),152155 flavonoids,156 vitamins C and E157 and coenzyme Q10 (CoQ10) have been tested in rodent models, although many of these agents had other roles in addition to being ROS scavengers (Table 1). Pro-longevity effects of antioxidants in the female reproductive system include maintenance of ovarian reserve, varying improvements of primordial and healthy ovarian follicle counts, a decreased proportion of atretic ovarian follicles and improvement in litter size and estrous cycle regularity. However, the use of antioxidants has also been associated with side effects, including those related to female reproduction, such as long-term disruption of ovarian and uterine functions with pharmacological doses of vitamins C and E.158 In fact, antioxidants such as vitamins A and E were shown to increase mortality in clinical trials.159 This may be explained by potential beneficial functions of oxidative stress in some contexts, for example, reduction of insulin resistance triggered by exercise-induced ROS.160 Additionally, several systematic reviews failed to demonstrate any positive outcome of antioxidant supplementation in the context of general aging159 and other age-related diseases, such as cataract, dementia and CVD.161164 In the context of ovarian aging, the in vivo studies on antioxidants were almost exclusively done in relatively young or middle-aged animals and failed to assess the effects in more aged ovaries (from older animals), potentially limiting the translatability to older or even postmenopausal women. Furthermore, only a few antioxidants have been studied in women with age-related ART failure and the results remain highly inconsistent.165,166 The oral supplementation of some antioxidants such as resveratrol was even associated with decreased pregnancy rates, the converse of what was expected.167 Reservatrol also has many aging-associated actions that are independent of its antioxidative effects,168 further complicating the interpretation of these findings.

Manipulation of glucose metabolism to mimic the effects of caloric restriction (CR) has been a focus in longevity studies. CR, the chronic reduction in total calorie intake without malnutrition, robustly improves both healthspan and lifespan in many organisms.169 As CR is not easy to be implemented in real life, genes and pathways involved in longevity mechanisms of CR are being identified and there are multiple candidate compounds that can potentially mimic the longevity effects of CR. Metformin, an approved anti-diabetic drug, achieves a reduction in blood glucose by enhancing peripheral insulin sensitivity and suppressing gluconeogenesis in the liver,170,171 which resemble some metabolic effects of CR.172 Indeed, metformin has demonstrated promising results in enhancing healthspan and lifespan in animal models172,173 and clinical trials.174 In the context of ovarian aging, multiple studies revealed that CR preserved oocyte quality, fertility and/or ovarian reserve in aged female rodents.6,175,176 Similarly, a recent study reported that six months of metformin treatment increased serum estrogen level and follicle quantity and resulted in more regular estrous cycles in normally aged mice.177 Such effects are consistent with another study showing an association between fasting-induced lower blood glucose level and reduced primordial follicular activation in mice.178 However, this observation was not corroborated by another rodent study,179 which found comparable follicle counts between metformin-fed rats and control rats. As there were differences in duration of treatment and rodent species, further investigations are needed to assess the effects of metformin in mammalian ovaries. Importantly, older animals need to be used to examine the true biological effects, as the previous studies were conducted on young rodents. These limitations apply to another study investigating 2-deoxyglucose (2-DG), where young mice and a short treatment duration were employed.180 2-DG is a synthetic glucose analog that competitively inhibits glycolysis, reduces insulin levels and decreases body temperature in rats, and thus has also been considered as a candidate CR mimetic.181 Interestingly, this study180 revealed inhibition of primordial follicle activation by 2-DG, indicating its potential in preservation of primordial ovarian follicles — hence protecting the ovarian reserve. However, the chronic treatment of 2-DG has been shown to cause cardiotoxicity and increased mortality in male rodents.182 Although it remains unknown whether this is also the case in females, such toxicity has decelerated the transition of 2-DG to clinical use.

Mitochondrial dysfunction has been identified as a hallmark for aging and implicated in ovarian aging and infertility, as oocytes are uniquely enriched with mitochondria.183 Aging-related changes in mitochondria, including accumulation of mitochondrial DNA mutations, altered membrane potential and impaired metabolism, undermine mitochondrial functions and are proposed to link to ovarian aging phenotypes.184 Besides the aforementioned antioxidant therapies, such as CoQ10, a combination of vitamins C and E, and flavonoids, aimed to increase nicotinamide adenine dinucleotide (NAD+), an essential cofactor and enzyme substrate in several crucial redox reactions and metabolic pathways which declines with age,185 has been shown to alleviate ovarian aging by improving mitochondrial function. NAD+ supplementation was tested in two recent studies in young and middle-aged mice.186,187 These studies demonstrated that the oocytes were rejuvenated, with enhanced fertility attributable to a reduction in levels of ROS and improvement in ovarian mitochondrial metabolism.186 However, knowledge about mitochondrial boosters including NAD+ precursors remain limited as they were only tested in rodents and in ages up to 14 months in mice. Therefore, mitochondrial boosters are likely to gain more attention in the future, especially given the new findings linking mitochondrial dysfunction to female reproductive aging via impaired NADH/NAD+ redox functions.188 Notably, the sirtuins, a family of NAD+-dependent deacylases and key regulators of aging, have both mitochondrial and non-mitochondrial functions such as DNA repair and inflammatory response.189 Thus, the mechanism of NAD+ supplementation in ovarian longevity may be pleiotropic.

mTOR is a serine/threonine protein complex that is sensitive to rapamycin and mTOR suppression has been shown to extend lifespan in several species.190199 mTOR also coordinates several key cellular signaling and metabolic pathways implicated in follicular development and ovarian aging.200 Follicular mTOR signaling stimulates primordial follicle activation, which is the start of post-puberty follicular development and directly determines the follicular reserve and reproductive lifespan. It was demonstrated in vivo that ovarian mTOR overactivation triggers premature follicular activation and early follicle depletion,201205 together with more atretic follicles and degenerated oocytes.205 At the same time, AKT-mediated mTOR signaling was found to regulate granulosa cell autophagy in folliculogenesis and its inhibition was found to induce follicular atresia through promoting granulosa cell autophagy.206 Studies in rodent models4,207210 have demonstrated that inhibition of mTOR signaling can improve ovarian reserve, as indicated by the increase in primordial follicle counts and extension of reproductive lifespan. There has also been some evidence indicating a decrease in the absolute atretic follicle count,210,211 probably due to overall suppression of follicular activation. Unfortunately, rapamycin was also found to cause disruption of estrous cycles and loss of fertility, due to a cessation of follicle activation after prolonged use of more than 4 months. This was expected because mTOR signaling plays a strong role in follicular growth as discussed. Nevertheless, this disruption seems to be reversible, as a recent study4 devised a 2-week transient rapamycin treatment that successfully restored follicular development and estrus cycles in post-treatment mice, observing an improvement in the treated mice’s reproductive capacity and ovarian lifespan, regardless of their ages at treatment up to 16 months of age. This approach may be consistent with the longevity effects of rapamycin, where a transient treatment even in relatively late age is sufficient to extend healthspan and lifespan.211 mTOR inhibitors could potentially be useful in the treatment of reproductive aging, but further studies are required due to considerations on utilizing mTOR inhibitors as strategic short-term treatment modality to achieve long-term protective effects against ovarian aging in women. Apart from inhibition, as mTOR plays a crucial role in reproduction, its chronic activation in reproductive aging should be extensively examined with small molecules such as MHY1485212 and 3BDO,213 which are potent mTOR activators.

Other options such as dehydroepiandrosterone (DHEA), a precursor for estrogen in peripheral tissues, and melatonin, a sleep-promoting hormone with antioxidant properties, have shown some success in extending reproductive lifespan and/or improving ovarian responses in both animal and clinical studies. The concentration of DHEA decreases progressively with age.214 As an essential prohormone in ovarian steroidogenesis, DHEA was suggested to promote gonadotropin action and ovarian functions, as indicated by the improvement in anti-Mullerian hormone (AMH) level and IVF outcomes in women with diminished ovarian reserve.215,216 This is further supported by an ovine study that also showed an increase in follicular AMH expression and granulosa cell proliferation after DHEA treatment.217 Yet, the same study also found an accelerated primordial follicle activation and an increase in antral follicle count, which raises the concern of pre-mature follicle depletion especially upon long-term administration. In contrast, melatonin, also an endogenous hormone in the body, has more extensive evidence in delaying ovarian aging. In rodents, it was found to improve oocyte quality and quantity and maintain follicular reserve.217224 A recent study223 demonstrated the endogenous role of melatonin in suppressing follicular overactivation and atresia and delaying ovarian aging. It was proposed that melatonin acts through its antioxidant capacity and the MT1/AMPK pathway,220,221,223 though further mechanistic study is needed. However, these studies varied greatly in age of subjects, end-point measurements and duration of treatment. For instance, supplementation of melatonin was conducted mostly on very young mice daily for half to one year,218,220222 which makes the translation of this treatment in clinical settings impractical due to the widespread metabolic and physiological activities that melatonin exerts on the body, raising concerns on any unanticipated adverse effects which can occur due to prolonged administration. Additionally, several clinical trials investigating oral melatonin administration in women with infertility or IVF failures have demonstrated conflicting results, which are summarized in Table 1.224229

A list of compounds tested in this context are listed in Table 1. However, many of the studies are primarily observational and although many compounds are purported to “extend reproductive lifespan”, results were often derived from findings in relatively young animals treated for varying durations. Furthermore, the exact mechanisms of actions for these interventions still lack clarity, and the therapeutic targets as well as their sites of action are yet to be elucidated. There remains a lot to be studied in understanding the underlying biological mechanisms governing reproductive aging at the ovarian follicle level and in the surrounding ovarian environment.

Future directions

There is a compelling need to develop new strategies to offer women to:

  1. Choose when she can fulfill her hopes of childbearing, once the mysteries behind ovarian senescence are better understood.

  2. Protect and enhance her reproductive lifespan and healthspan using innovative solutions that are safe, with minimal or no side effects, to overcome the negative consequences of reproductive aging.

  3. Advocate for reproductive longevity and equality in women, and for science to assist in tackling the inevitability of reproductive aging for future generations.

New intervention strategies should target the root cause of the reproductive aging process, ideally at a specific organ-cell-molecular target level, and be relatively short-term and have minimal side effects.

Conclusion

Current knowledge on the mechanisms of ovarian or reproductive aging remains limited. Clinical management of fertility issues is always confounded by maternal age with reproductive outcomes limited by the age of women, i.e., older age and reduced ovarian reserves result in higher risks of reproductive failure.230 Few options are available for the management of women with POI and natural menopause because we still do not fully understand the biology of ovarian senescence in women. The balance between reproductive lifespan (fertility) and healthspan (general good health) will be the long-term goal and imperative as part of the movement for gender equality. A woman should not be limited to her reproductive lifespan and “accept” what biology has imposed on her reproductive choices and life choices, in addition to her decline of healthspan later in life. Women represent half of the world population, and they also carry the future of the world. Reproductive longevity in women also determines healthy longevity in their male relatives,231 which emphasizes the importance of solving female reproductive senescence and will open doors to discover ways to enhance true healthy longevity for both men and women.

Acknowledgements

L.D. is funded for her PhD studies by National University of Singapore. The authors acknowledge the generous contribution of the Bia-Echo Foundation to Yong Loo Lin School of Medicine, National University of Singapore in the set-up of NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality to change the science and narrative of female reproductive longevity.

Author contributions

Z.H. and B.K.K. conceptualized and led the review. D.B.L.T. and L.D. performed the literature review, summarized data, constructed the figures and wrote the first version of the manuscript. B.K.K. and Z.H. revised and critically reviewed the subsequent draft revisions. All authors read, edited, and approved the final article.

Competing interests

The authors declare no competing interests.

Footnotes

These authors contributed equally: Lu Dong, Daniel Boon Loong Teh.

Contributor Information

Brian Keith Kennedy, Email: bkennedy@nus.edu.sg.

Zhongwei Huang, Email: obgzwh@nus.edu.sg.

References

  • 1.United Nations Department of Economic and Social Affairs. World Mortality 2019 Data Booklet (United Nations Department of Economic and Social Affairs, 2019).
  • 2.Ginter E, Simko V. Women live longer than men. Bratisl. Lek. Listy. 2013;114:45–49. doi: 10.4149/bll_2013_011. [DOI] [PubMed] [Google Scholar]
  • 3.Li Q, et al. Current understanding of ovarian aging. Sci. China Life Sci. 2012;55:659–669. doi: 10.1007/s11427-012-4352-5. [DOI] [PubMed] [Google Scholar]
  • 4.Dou X, et al. Short-term rapamycin treatment increases ovarian lifespan in young and middle-aged female mice. Aging Cell. 2017;16:825–836. doi: 10.1111/acel.12617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zhang J, et al. Can ovarian aging be delayed by pharmacological strategies? Aging. 2019;11:817–832. doi: 10.18632/aging.101784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Garcia DN, et al. Effect of caloric restriction and rapamycin on ovarian aging in mice. Geroscience. 2019;41:395–408. doi: 10.1007/s11357-019-00087-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Faddy MJ, Gosden RG, Gougeon A, Richardson SJ, Nelson JF. Accelerated disappearance of ovarian follicles in mid-life: implications for forecasting menopause. Hum. Reprod. 1992;7:1342–1346. doi: 10.1093/oxfordjournals.humrep.a137570. [DOI] [PubMed] [Google Scholar]
  • 8.Broekmans FJ, Soules MR, Fauser BC. Ovarian aging: mechanisms and clinical consequences. Endocr. Rev. 2009;30:465–493. doi: 10.1210/er.2009-0006. [DOI] [PubMed] [Google Scholar]
  • 9.Santoro N. The menopausal transition. Am. J. Med. 2005;118(Suppl 12B):8–13. doi: 10.1016/j.amjmed.2005.09.008. [DOI] [PubMed] [Google Scholar]
  • 10.Laven JS. Genetics of early and normal menopause. Semin. Reprod. Med. 2015;33:377–383. doi: 10.1055/s-0035-1567825. [DOI] [PubMed] [Google Scholar]
  • 11.Zhu D, Li X, Macrae VE, Simoncini T, Fu X. Extragonadal effects of follicle-stimulating hormone on osteoporosis and cardiovascular disease in women during menopausal transition. Trends Endocrinol. Metab. 2018;29:571–580. doi: 10.1016/j.tem.2018.06.001. [DOI] [PubMed] [Google Scholar]
  • 12.Lizcano F, Guzman G. Estrogen deficiency and the origin of obesity during menopause. Biomed. Res. Int. 2014;2014:757461. doi: 10.1155/2014/757461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Song X, et al. Reproductive and hormonal factors and risk of cognitive impairment among Singapore Chinese women. Am. J. Obstet. Gynecol. 2020;223:410.e411–410.e423. doi: 10.1016/j.ajog.2020.02.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Muka T, et al. Association of age at onset of menopause and time since onset of menopause with cardiovascular outcomes, intermediate vascular traits, and all-cause mortality: a systematic review and meta-analysis. JAMA Cardiol. 2016;1:767–776. doi: 10.1001/jamacardio.2016.2415. [DOI] [PubMed] [Google Scholar]
  • 15.Thong EP, Codner E, Laven JSE, Teede H. Diabetes: a metabolic and reproductive disorder in women. Lancet Diabetes Endocrinol. 2020;8:134–149. doi: 10.1016/S2213-8587(19)30345-6. [DOI] [PubMed] [Google Scholar]
  • 16.Geraci A, et al. Sarcopenia and menopause: the role of estradiol. Front. Endocrinol. 2021;12:682012. doi: 10.3389/fendo.2021.682012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhu D, et al. Age at natural menopause and risk of incident cardiovascular disease: a pooled analysis of individual patient data. Lancet Public Health. 2019;4:e553–e564. doi: 10.1016/S2468-2667(19)30155-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Scheffer GJ, et al. The number of antral follicles in normal women with proven fertility is the best reflection of reproductive age. Hum. Reprod. 2003;18:700–706. doi: 10.1093/humrep/deg135. [DOI] [PubMed] [Google Scholar]
  • 19.Drummond AE. The role of steroids in follicular growth. Reprod. Biol. Endocrinol. 2006;4:16–16. doi: 10.1186/1477-7827-4-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Khosla S, Oursler MJ, Monroe DG. Estrogen and the skeleton. Trends Endocrinol. Metab. 2012;23:576–581. doi: 10.1016/j.tem.2012.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Russell JK, Jones CK, Newhouse PA. The role of estrogen in brain and cognitive aging. Neurotherapeutics. 2019;16:649–665. doi: 10.1007/s13311-019-00766-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wilkinson HN, Hardman MJ. The role of estrogen in cutaneous ageing and repair. Maturitas. 2017;103:60–64. doi: 10.1016/j.maturitas.2017.06.026. [DOI] [PubMed] [Google Scholar]
  • 23.Knowlton AA, Lee AR. Estrogen and the cardiovascular system. Pharmacol. Ther. 2012;135:54–70. doi: 10.1016/j.pharmthera.2012.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sitruk-Ware R, El-Etr M. Progesterone and related progestins: potential new health benefits. Climacteric. 2013;16:69–78. doi: 10.3109/13697137.2013.802556. [DOI] [PubMed] [Google Scholar]
  • 25.Gleicher N, Weghofer A, Barad DH. Defining ovarian reserve to better understand ovarian aging. Reprod. Biol. Endocrinol. 2011;9:23–23. doi: 10.1186/1477-7827-9-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mesiano, S., Jones, E. E. Chapter 55: The female reproductive system. In: Medical physiology 3rd ed. (eds Boron, W., Boulpaep, E.) (Philadelphia, Elsevier, 2017).
  • 27.Pellestor F, Anahory T, Hamamah S. Effect of maternal age on the frequency of cytogenetic abnormalities in human oocytes. Cytogenet. Genome Res. 2005;111:206–212. doi: 10.1159/000086891. [DOI] [PubMed] [Google Scholar]
  • 28.Kuliev A, Cieslak J, Verlinsky Y. Frequency and distribution of chromosome abnormalities in human oocytes. Cytogenet. Genome Res. 2005;111:193–198. doi: 10.1159/000086889. [DOI] [PubMed] [Google Scholar]
  • 29.Battaglia DE, Goodwin P, Klein NA, Soules MR. Influence of maternal age on meiotic spindle assembly in oocytes from naturally cycling women. Hum. Reprod. 1996;11:2217–2222. doi: 10.1093/oxfordjournals.humrep.a019080. [DOI] [PubMed] [Google Scholar]
  • 30.Liu Y, et al. Age-related changes in the mitochondria of human mural granulosa cells. Hum. Reprod. 2017;32:2465–2473. doi: 10.1093/humrep/dex309. [DOI] [PubMed] [Google Scholar]
  • 31.Seifer DB, DeJesus V, Hubbard K. Mitochondrial deletions in luteinized granulosa cells as a function of age in women undergoing in vitro fertilization. Fertil. Steril. 2002;78:1046–1048. doi: 10.1016/S0015-0282(02)04214-0. [DOI] [PubMed] [Google Scholar]
  • 32.Sreerangaraja Urs DB, et al. Mitochondrial function in modulating human granulosa cell steroidogenesis and female fertility. Int. J. Mol. Sci. 2020;21:3592. doi: 10.3390/ijms21103592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Tatone C, et al. Evidence that carbonyl stress by methylglyoxal exposure induces DNA damage and spindle aberrations, affects mitochondrial integrity in mammalian oocytes and contributes to oocyte ageing. Hum. Reprod. 2011;26:1843–1859. doi: 10.1093/humrep/der140. [DOI] [PubMed] [Google Scholar]
  • 34.Chang Y, Li J, Li X, Liu H, Liang X. Egg quality and pregnancy outcome in young infertile women with diminished ovarian reserve. Med. Sci. Monit. 2018;24:7279–7284. doi: 10.12659/MSM.910410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Pollycove R, Naftolin F, Simon JA. The evolutionary origin and significance of menopause. Menopause. 2011;18:336–342. doi: 10.1097/gme.0b013e3181ed957a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ellis S, et al. Postreproductive lifespans are rare in mammals. Ecol. Evol. 2018;8:2482–2494. doi: 10.1002/ece3.3856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hawkes K. Human longevity: the grandmother effect. Nature. 2004;428:128–129. doi: 10.1038/428128a. [DOI] [PubMed] [Google Scholar]
  • 38.Hawkes K, O’Connell JF, Jones NG, Alvarez H, Charnov EL. Grandmothering, menopause, and the evolution of human life histories. Proc. Natl. Acad. Sci. USA. 1998;95:1336–1339. doi: 10.1073/pnas.95.3.1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Whitehead H. Life history evolution: what does a menopausal killer whale do? Curr. Biol. 2015;25:R225–R227. doi: 10.1016/j.cub.2015.02.002. [DOI] [PubMed] [Google Scholar]
  • 40.Singh A, Kaur S, Walia I. A historical perspective on menopause and menopausal age. Bull. Indian Inst. Hist. Med. Hyderabad. 2002;32:121–135. [PubMed] [Google Scholar]
  • 41.Velez MP, et al. Age at natural menopause and physical functioning in postmenopausal women: the Canadian Longitudinal Study on Aging. Menopause. 2019;26:958–965. doi: 10.1097/GME.0000000000001362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Vasold M. [Mortality in Nuremberg in the 19th century (about 1800 to 1913)] Wurzbg Medizinhist Mitt. 2006;25:241–338. [PubMed] [Google Scholar]
  • 43.Anderson RN. Deaths: leading causes for 2000. Natl. Vital Stat. Rep. 2002;50:1–85. [PubMed] [Google Scholar]
  • 44.Shaw-Taylor L. An introduction to the history of infectious diseases, epidemics and the early phases of the long-run decline in mortality. Econ. Hist. Rev. 2020;73:E1–E19. doi: 10.1111/ehr.13019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Oeppen J, Vaupel JW. Demography. Broken limits to life expectancy. Science. 2002;296:1029–1031. doi: 10.1126/science.1069675. [DOI] [PubMed] [Google Scholar]
  • 46.Xirocostas ZA, Everingham SE, Moles AT. The sex with the reduced sex chromosome dies earlier: a comparison across the tree of life. Biol. Lett. 2020;16:20190867. doi: 10.1098/rsbl.2019.0867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hawkes K, Smith KR. Do women stop early? Similarities in fertility decline in humans and chimpanzees. Ann. N. Y. Acad. Sci. 2010;1204:43–53. doi: 10.1111/j.1749-6632.2010.05527.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wood BM, Watts DP, Mitani JC, Langergraber KE. Favorable ecological circumstances promote life expectancy in chimpanzees similar to that of human hunter-gatherers. J. Hum. Evol. 2017;105:41–56. doi: 10.1016/j.jhevol.2017.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Shadyab AH, et al. Ages at menarche and menopause and reproductive lifespan as predictors of exceptional longevity in women: the Women’s Health Initiative. Menopause. 2017;24:35–44. doi: 10.1097/GME.0000000000000710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hodson L, et al. Lower resting and total energy expenditure in postmenopausal compared with premenopausal women matched for abdominal obesity. J. Nutr. Sci. 2014;3:e3. doi: 10.1017/jns.2013.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Vincent-Rohfritsch A, Le Ray C, Anselem O, Cabrol D, Goffinet F. Pregnancy in women aged 43 years or older: maternal and perinatal risks. J. Gynecol. Obstet. Biol. Reprod. 2012;41:468–475. doi: 10.1016/j.jgyn.2012.04.013. [DOI] [PubMed] [Google Scholar]
  • 52.Yogev Y, et al. Pregnancy outcome at extremely advanced maternal age. Am. J. Obstet. Gynecol. 2010;203:558.e1–558.e7. doi: 10.1016/j.ajog.2010.07.039. [DOI] [PubMed] [Google Scholar]
  • 53.Sheen JJ, et al. Maternal age and risk for adverse outcomes. Am. J. Obstet. Gynecol. 2018;219:390.e1–390.e15. doi: 10.1016/j.ajog.2018.08.034. [DOI] [PubMed] [Google Scholar]
  • 54.Londero AP, Rossetti E, Pittini C, Cagnacci A, Driul L. Maternal age and the risk of adverse pregnancy outcomes: a retrospective cohort study. BMC Pregnancy Childbirth. 2019;19:261. doi: 10.1186/s12884-019-2400-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mikwar M, MacFarlane AJ, Marchetti F. Mechanisms of oocyte aneuploidy associated with advanced maternal age. Mutat. Res. 2020;785:108320. doi: 10.1016/j.mrrev.2020.108320. [DOI] [PubMed] [Google Scholar]
  • 56.Babiker FA, et al. 17beta-estradiol antagonizes cardiomyocyte hypertrophy by autocrine/paracrine stimulation of a guanylyl cyclase A receptor-cyclic guanosine monophosphate-dependent protein kinase pathway. Circulation. 2004;109:269–276. doi: 10.1161/01.CIR.0000105682.85732.BD. [DOI] [PubMed] [Google Scholar]
  • 57.Lagranha CJ, Deschamps A, Aponte A, Steenbergen C, Murphy E. Sex differences in the phosphorylation of mitochondrial proteins result in reduced production of reactive oxygen species and cardioprotection in females. Circ. Res. 2010;106:1681–1691. doi: 10.1161/CIRCRESAHA.109.213645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Iorga A, et al. Rescue of pressure overload-induced heart failure by estrogen therapy. J. Am. Heart Assoc. 2016;5:e002482. doi: 10.1161/JAHA.115.002482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Pedram A, Razandi M, Narayanan R, Levin ER. Estrogen receptor beta signals to inhibition of cardiac fibrosis. Mol. Cell Endocrinol. 2016;434:57–68. doi: 10.1016/j.mce.2016.06.018. [DOI] [PubMed] [Google Scholar]
  • 60.Adams MR, et al. Inhibition of coronary artery atherosclerosis by 17-beta estradiol in ovariectomized monkeys. Lack of an effect of added progesterone. Arteriosclerosis. 1990;10:1051–1057. doi: 10.1161/01.ATV.10.6.1051. [DOI] [PubMed] [Google Scholar]
  • 61.Christ JP, et al. Estrogen deprivation and cardiovascular disease risk in primary ovarian insufficiency. Fertil. Steril. 2018;109:594–600.e1. doi: 10.1016/j.fertnstert.2017.11.035. [DOI] [PubMed] [Google Scholar]
  • 62.Giordano S, et al. Estrogen and cardiovascular disease: is timing everything? Am. J. Med. Sci. 2015;350:27–35. doi: 10.1097/MAJ.0000000000000512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Spence RD, Voskuhl RR. Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration. Front. Neuroendocrinol. 2012;33:105–115. doi: 10.1016/j.yfrne.2011.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Cooper GS, Stroehla BC. The epidemiology of autoimmune diseases. Autoimmun. Rev. 2003;2:119–125. doi: 10.1016/S1568-9972(03)00006-5. [DOI] [PubMed] [Google Scholar]
  • 65.DECODE Study Group. Age- and sex-specific prevalences of diabetes and impaired glucose regulation in 13 European cohorts. Diabetes Care. 2003;26:61–69. doi: 10.2337/diacare.26.1.61. [DOI] [PubMed] [Google Scholar]
  • 66.Qiao Q, et al. Age- and sex-specific prevalence of diabetes and impaired glucose regulation in 11 Asian cohorts. Diabetes Care. 2003;26:1770–1780. doi: 10.2337/diacare.26.6.1770. [DOI] [PubMed] [Google Scholar]
  • 67.Gubbels Bupp MR. Sex, the aging immune system, and chronic disease. Cell Immunol. 2015;294:102–110. doi: 10.1016/j.cellimm.2015.02.002. [DOI] [PubMed] [Google Scholar]
  • 68.Vitale C, Mendelsohn ME, Rosano GMC. Gender differences in the cardiovascular effect of sex hormones. Nat. Rev. Cardiol. 2009;6:532–542. doi: 10.1038/nrcardio.2009.105. [DOI] [PubMed] [Google Scholar]
  • 69.Lerner DJ, Kannel WB. Patterns of coronary heart disease morbidity and mortality in the sexes: a 26-year follow-up of the Framingham population. Am. Heart J. 1986;111:383–390. doi: 10.1016/0002-8703(86)90155-9. [DOI] [PubMed] [Google Scholar]
  • 70.Pennell LM, Galligan CL, Fish EN. Sex affects immunity. J. Autoimmun. 2012;38:J282–J291. doi: 10.1016/j.jaut.2011.11.013. [DOI] [PubMed] [Google Scholar]
  • 71.Ghisletti S, Meda C, Maggi A, Vegeto E. 17beta-estradiol inhibits inflammatory gene expression by controlling NF-kappaB intracellular localization. Mol. Cell Biol. 2005;25:2957–2968. doi: 10.1128/MCB.25.8.2957-2968.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Rettew JA, Huet YM, Marriott I. Estrogens augment cell surface TLR4 expression on murine macrophages and regulate sepsis susceptibility in vivo. Endocrinology. 2009;150:3877–3884. doi: 10.1210/en.2009-0098. [DOI] [PubMed] [Google Scholar]
  • 73.Rettew JA, McCall SHT, Marriott I. GPR30/GPER-1 mediates rapid decreases in TLR4 expression on murine macrophages. Mol. Cell Endocrinol. 2010;328:87–92. doi: 10.1016/j.mce.2010.07.017. [DOI] [PubMed] [Google Scholar]
  • 74.Chidi-Ogbolu N, Baar K. Effect of estrogen on musculoskeletal performance and injury risk. Front. Physiol. 2018;9:1834. doi: 10.3389/fphys.2018.01834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Luo T, Kim JK. The role of estrogen and estrogen receptors on cardiomyocytes: an overview. Can. J. Cardiol. 2016;32:1017–1025. doi: 10.1016/j.cjca.2015.10.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Keefe AC, et al. Muscle stem cells contribute to myofibres in sedentary adult mice. Nat. Commun. 2015;6:7087. doi: 10.1038/ncomms8087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Collins BC, et al. Estrogen regulates the satellite cell compartment in females. Cell Rep. 2019;28:368–381.e6. doi: 10.1016/j.celrep.2019.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Shultz SJ, Sander TC, Kirk SE, Perrin DH. Sex differences in knee joint laxity change across the female menstrual cycle. J. Sports Med. Phys. Fitness. 2005;45:594–603. [PMC free article] [PubMed] [Google Scholar]
  • 79.Fischer V, Haffner-Luntzer M. Interaction between bone and immune cells: implications for postmenopausal osteoporosis. Semin. Cell Dev. Biol. 2022;123:14–21. doi: 10.1016/j.semcdb.2021.05.014. [DOI] [PubMed] [Google Scholar]
  • 80.Krum SA, Brown M. Unraveling estrogen action in osteoporosis. Cell Cycle. 2008;7:1348–1352. doi: 10.4161/cc.7.10.5892. [DOI] [PubMed] [Google Scholar]
  • 81.Mehta J, Kling JM, Manson JE. Risks, benefits, and treatment modalities of menopausal hormone therapy: current concepts. Front. Endocrinol. 2021;12:564781. doi: 10.3389/fendo.2021.564781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Cauley JA. Estrogen and bone health in men and women. Steroids. 2015;99:11–15. doi: 10.1016/j.steroids.2014.12.010. [DOI] [PubMed] [Google Scholar]
  • 83.Borjesson AE, Lagerquist MK, Windahl SH, Ohlsson C. The role of estrogen receptor alpha in the regulation of bone and growth plate cartilage. Cell Mol. Life Sci. 2013;70:4023–4037. doi: 10.1007/s00018-013-1317-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Krum SA, et al. Estrogen protects bone by inducing Fas ligand in osteoblasts to regulate osteoclast survival. EMBO J. 2008;27:535–545. doi: 10.1038/sj.emboj.7601984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hughes DE, et al. Estrogen promotes apoptosis of murine osteoclasts mediated by TGF-beta. Nat. Med. 1996;2:1132–1136. doi: 10.1038/nm1096-1132. [DOI] [PubMed] [Google Scholar]
  • 86.Almeida M, et al. Estrogen receptor-alpha signaling in osteoblast progenitors stimulates cortical bone accrual. J. Clin. Invest. 2013;123:394–404. doi: 10.1172/JCI65910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Raisz LG. Pathogenesis of osteoporosis: concepts, conflicts, and prospects. J. Clin. Invest. 2005;115:3318–3325. doi: 10.1172/JCI27071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Streicher C, et al. Estrogen regulates bone turnover by targeting RANKL expression in bone lining cells. Sci. Rep. 2017;7:6460. doi: 10.1038/s41598-017-06614-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Yousefzadeh N, Kashfi K, Jeddi S, Ghasemi A. Ovariectomized rat model of osteoporosis: a practical guide. EXCLI J. 2020;19:89–107. doi: 10.17179/excli2019-1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Onal M, et al. Receptor activator of nuclear factor kappaB ligand (RANKL) protein expression by B lymphocytes contributes to ovariectomy-induced bone loss. J. Biol. Chem. 2012;287:29851–29860. doi: 10.1074/jbc.M112.377945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Mohammad I, et al. Estrogen receptor alpha contributes to T cell-mediated autoimmune inflammation by promoting T cell activation and proliferation. Sci. Signal. 2018;11:eaap9415. doi: 10.1126/scisignal.aap9415. [DOI] [PubMed] [Google Scholar]
  • 92.Fuller K, Murphy C, Kirstein B, Fox SW, Chambers TJ. TNFalpha potently activates osteoclasts, through a direct action independent of and strongly synergistic with RANKL. Endocrinology. 2002;143:1108–1118. doi: 10.1210/endo.143.3.8701. [DOI] [PubMed] [Google Scholar]
  • 93.Weitzmann MN, Ofotokun I. Physiological and pathophysiological bone turnover — role of the immune system. Nat. Rev. Endocrinol. 2016;12:518–532. doi: 10.1038/nrendo.2016.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Vannucci L, et al. Calcium intake in bone health: a focus on calcium-rich mineral waters. Nutrients. 2018;10:1930. doi: 10.3390/nu10121930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Christakos S, Dhawan P, Porta A, Mady LJ, Seth T. Vitamin D and intestinal calcium absorption. Mol. Cell Endocrinol. 2011;347:25–29. doi: 10.1016/j.mce.2011.05.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gallagher JC. Vitamin D and aging. Endocrinol. Metab. Clin. North Am. 2013;42:319–332. doi: 10.1016/j.ecl.2013.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Veldurthy V, et al. Vitamin D, calcium homeostasis and aging. Bone Res. 2016;4:16041. doi: 10.1038/boneres.2016.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Tella SH, Gallagher JC. Prevention and treatment of postmenopausal osteoporosis. J. Steroid Biochem. Mol. Biol. 2014;142:155–170. doi: 10.1016/j.jsbmb.2013.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Anderson GL, et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA. 2004;291:1701–1712. doi: 10.1001/jama.291.14.1701. [DOI] [PubMed] [Google Scholar]
  • 100.Rossouw JE, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321–333. doi: 10.1001/jama.288.3.321. [DOI] [PubMed] [Google Scholar]
  • 101.Levin VA, Jiang X, Kagan R. Estrogen therapy for osteoporosis in the modern era. Osteoporos Int. 2018;29:1049–1055. doi: 10.1007/s00198-018-4414-z. [DOI] [PubMed] [Google Scholar]
  • 102.Hulley S, et al. Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA. 1998;280:605–613. doi: 10.1001/jama.280.7.605. [DOI] [PubMed] [Google Scholar]
  • 103.Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc. Natl. Acad. Sci. USA. 2000;97:12729–12734. doi: 10.1073/pnas.97.23.12729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Murata Y, Robertson KM, Jones ME, Simpson ER. Effect of estrogen deficiency in the male: the ArKO mouse model. Mol. Cell Endocrinol. 2002;193:7–12. doi: 10.1016/S0303-7207(02)00090-4. [DOI] [PubMed] [Google Scholar]
  • 105.Grodstein F, et al. A prospective, observational study of postmenopausal hormone therapy and primary prevention of cardiovascular disease. Ann. Intern. Med. 2000;133:933–941. doi: 10.7326/0003-4819-133-12-200012190-00008. [DOI] [PubMed] [Google Scholar]
  • 106.Miller WL, Auchus RJ. The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr. Rev. 2011;32:81–151. doi: 10.1210/er.2010-0013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Rodriguez-Cuenca S, et al. Sex steroid receptor expression profile in brown adipose tissue. Effects of hormonal status. Cell Physiol. Biochem. 2007;20:877–886. doi: 10.1159/000110448. [DOI] [PubMed] [Google Scholar]
  • 108.Chen HJ, Meng T, Gao PJ, Ruan CC. The role of brown adipose tissue dysfunction in the development of cardiovascular disease. Front. Endocrinol. 2021;12:652246. doi: 10.3389/fendo.2021.652246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Planavila A, et al. Fibroblast growth factor 21 protects against cardiac hypertrophy in mice. Nat. Commun. 2013;4:2019. doi: 10.1038/ncomms3019. [DOI] [PubMed] [Google Scholar]
  • 110.Hondares E, et al. Thermogenic activation induces FGF21 expression and release in brown adipose tissue. J. Biol. Chem. 2011;286:12983–12990. doi: 10.1074/jbc.M110.215889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Park CJ, et al. Genetic rescue of nonclassical ERalpha signaling normalizes energy balance in obese Eralpha-null mutant mice. J. Clin. Invest. 2011;121:604–612. doi: 10.1172/JCI41702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Casazza K, Page GP, Fernandez JR. The association between the rs2234693 and rs9340799 estrogen receptor alpha gene polymorphisms and risk factors for cardiovascular disease: a review. Biol. Res. Nurs. 2010;12:84–97. doi: 10.1177/1099800410371118. [DOI] [PubMed] [Google Scholar]
  • 113.Schierbeck LL, et al. Effect of hormone replacement therapy on cardiovascular events in recently postmenopausal women: randomised trial. BMJ. 2012;345:e6409. doi: 10.1136/bmj.e6409. [DOI] [PubMed] [Google Scholar]
  • 114.Hodis HN, et al. Methods and baseline cardiovascular data from the Early versus Late Intervention Trial with Estradiol testing the menopausal hormone timing hypothesis. Menopause. 2015;22:391–401. doi: 10.1097/GME.0000000000000343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Harman SM, et al. Arterial imaging outcomes and cardiovascular risk factors in recently menopausal women: a randomized trial. Ann. Intern. Med. 2014;161:249–260. doi: 10.7326/M14-0353. [DOI] [PubMed] [Google Scholar]
  • 116.Phillips LS, Langer RD. Postmenopausal hormone therapy: critical reappraisal and a unified hypothesis. Fertil. Steril. 2005;83:558–566. doi: 10.1016/j.fertnstert.2004.11.012. [DOI] [PubMed] [Google Scholar]
  • 117.Cavero-Redondo I, Peleteiro B, Alvarez-Bueno C, Rodriguez-Artalejo F, Martinez-Vizcaino V. Glycated haemoglobin A1c as a risk factor of cardiovascular outcomes and all-cause mortality in diabetic and non-diabetic populations: a systematic review and meta-analysis. BMJ Open. 2017;7:e015949. doi: 10.1136/bmjopen-2017-015949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zhong GC, Ye MX, Cheng JH, Zhao Y, Gong JP. HbA1c and risks of all-cause and cause-specific death in subjects without known diabetes: a dose-response meta-analysis of prospective cohort studies. Sci. Rep. 2016;6:24071. doi: 10.1038/srep24071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Iorga A, et al. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol. Sex Differ. 2017;8:33. doi: 10.1186/s13293-017-0152-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yang XP, Reckelhoff JF. Estrogen, hormonal replacement therapy and cardiovascular disease. Curr. Opin. Nephrol. Hypertens. 2011;20:133–138. doi: 10.1097/MNH.0b013e3283431921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Zárate S, Stevnsner T, Gredilla R. Role of estrogen and other sex hormones in brain aging. neuroprotection and DNA repair. Front. Aging Neurosci. 2017;9:430–430. doi: 10.3389/fnagi.2017.00430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Siddiqui AN, et al. Neuroprotective role of steroidal sex hormones: an overview. CNS Neurosci. Ther. 2016;22:342–350. doi: 10.1111/cns.12538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Gambacciani M, Levancini M. Hormone replacement therapy and the prevention of postmenopausal osteoporosis. Prz Menopauzalny. 2014;13:213–220. doi: 10.5114/pm.2014.44996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Fait T. Menopause hormone therapy: latest developments and clinical practice. Drugs Context. 2019;8:212551–212551. doi: 10.7573/dic.212551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Vinogradova Y, Coupland C, Hippisley-Cox J. Use of hormone replacement therapy and risk of breast cancer: nested case-control studies using the QResearch and CPRD databases. BMJ. 2020;371:m3873. doi: 10.1136/bmj.m3873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Lobo RA. Hormone-replacement therapy: current thinking. Nat. Rev. Endocrinol. 2017;13:220–231. doi: 10.1038/nrendo.2016.164. [DOI] [PubMed] [Google Scholar]
  • 127.D’Alonzo M, Bounous VE, Villa M, Biglia N. Current evidence of the oncological benefit-risk profile of hormone replacement therapy. Medicina. 2019;55:573. doi: 10.3390/medicina55090573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Boardman HMP, et al. Hormone therapy for preventing cardiovascular disease in post‐menopausal women. Cochrane Database Syst. Rev. 2015;2015:CD002229. doi: 10.1002/14651858.CD002229.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lethaby A, Hogervorst E, Richards M, Yesufu A, Yaffe K. Hormone replacement therapy for cognitive function in postmenopausal women. Cochrane Database Syst. Rev. 2018;2008:CD003122. doi: 10.1002/14651858.CD003122.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Clegg A, Young J, Iliffe S, Rikkert MO, Rockwood K. Frailty in elderly people. Lancet. 2013;381:752–762. doi: 10.1016/S0140-6736(12)62167-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Jaul E, Barron J. Age-related diseases and clinical and public health implications for the 85 years old and over population. Front. Public Health. 2017;5:335–335. doi: 10.3389/fpubh.2017.00335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Cheng X, et al. Population ageing and mortality during 1990–2017: a global decomposition analysis. PLoS Med. 2020;17:e1003138. doi: 10.1371/journal.pmed.1003138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Olshansky SJ. From lifespan to healthspan. JAMA. 2018;320:1323–1324. doi: 10.1001/jama.2018.12621. [DOI] [PubMed] [Google Scholar]
  • 134.Crimmins EM. Lifespan and healthspan: past, present, and promise. Gerontologist. 2015;55:901–911. doi: 10.1093/geront/gnv130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Argyle CE, Harper JC, Davies MC. Oocyte cryopreservation: where are we now? Hum. Reprod. Update. 2016;22:440–449. doi: 10.1093/humupd/dmw007. [DOI] [PubMed] [Google Scholar]
  • 136.Iussig B, et al. A brief history of oocyte cryopreservation: arguments and facts. Acta Obstet. Gynecol. Scand. 2019;98:550–558. doi: 10.1111/aogs.13569. [DOI] [PubMed] [Google Scholar]
  • 137.Bhattacharya S, Maheshwari A, Mollison J. Factors associated with failed treatment: an analysis of 121,744 women embarking on their first IVF cycles. PLoS One. 2013;8:e82249. doi: 10.1371/journal.pone.0082249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Centers for Disease Control and Prevention. 2019 Assisted Reproductive Technology Fertility Clinic and National Summary Report (US Dept of Health and Human Services, 2021).
  • 139.Llarena N, Hine C. Reproductive longevity and aging: geroscience approaches to maintain long-term ovarian fitness. J. Gerontol. A Biol. Sci. Med. Sci. 2021;76:1551–1560. doi: 10.1093/gerona/glaa204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Finch CE. The menopause and aging, a comparative perspective. J. Steroid Biochem. Mol. Biol. 2014;142:132–141. doi: 10.1016/j.jsbmb.2013.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Harman D. Aging: a theory based on free radical and radiation chemistry. J. Gerontol. 1956;11:298–300. doi: 10.1093/geronj/11.3.298. [DOI] [PubMed] [Google Scholar]
  • 142.Liochev SI. Reactive oxygen species and the free radical theory of aging. Free Radic. Biol. Med. 2013;60:1–4. doi: 10.1016/j.freeradbiomed.2013.02.011. [DOI] [PubMed] [Google Scholar]
  • 143.Sohal RS, Weindruch R. Oxidative stress, caloric restriction, and aging. Science. 1996;273:59–63. doi: 10.1126/science.273.5271.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature. 2000;408:239–247. doi: 10.1038/35041687. [DOI] [PubMed] [Google Scholar]
  • 145.Sasaki H, et al. Impact of oxidative stress on age-associated decline in oocyte developmental competence. Front. Endocrinol. 2019;10:811–811. doi: 10.3389/fendo.2019.00811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Becatti M, et al. A biochemical approach to detect oxidative stress in infertile women undergoing assisted reproductive technology procedures. Int. J. Mol. Sci. 2018;19:592. doi: 10.3390/ijms19020592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wiener-Megnazi Z, et al. Oxidative stress indices in follicular fluid as measured by the thermochemiluminescence assay correlate with outcome parameters in in vitro fertilization. Fertil. Steril. 2004;82:1171–1176. doi: 10.1016/j.fertnstert.2004.06.013. [DOI] [PubMed] [Google Scholar]
  • 148.Appasamy M, et al. Evaluation of the relationship between follicular fluid oxidative stress, ovarian hormones, and response to gonadotropin stimulation. Fertil. Steril. 2008;89:912–921. doi: 10.1016/j.fertnstert.2007.04.034. [DOI] [PubMed] [Google Scholar]
  • 149.Oyawoye O, et al. Antioxidants and reactive oxygen species in follicular fluid of women undergoing IVF: relationship to outcome. Hum. Reprod. 2003;18:2270–2274. doi: 10.1093/humrep/deg450. [DOI] [PubMed] [Google Scholar]
  • 150.Das S, et al. Reactive oxygen species level in follicular fluid — embryo quality marker in IVF? Hum. Reprod. 2006;21:2403–2407. doi: 10.1093/humrep/del156. [DOI] [PubMed] [Google Scholar]
  • 151.Terao H, et al. Role of oxidative stress in follicular fluid on embryos of patients undergoing assisted reproductive technology treatment. J. Obstet. Gynaecol. Res. 2019;45:1884–1891. doi: 10.1111/jog.14040. [DOI] [PubMed] [Google Scholar]
  • 152.Liu L, Trimarchi JR, Navarro P, Blasco MA, Keefe DL. Oxidative stress contributes to arsenic-induced telomere attrition, chromosome instability, and apoptosis. J. Biol. Chem. 2003;278:31998–32004. doi: 10.1074/jbc.M303553200. [DOI] [PubMed] [Google Scholar]
  • 153.Navarro PA, Liu L, Ferriani RA, Keefe DL. Arsenite induces aberrations in meiosis that can be prevented by coadministration of N-acetylcysteine in mice. Fertil. Steril. 2006;85(Suppl 1):1187–1194. doi: 10.1016/j.fertnstert.2005.08.060. [DOI] [PubMed] [Google Scholar]
  • 154.Huang J, Okuka M, McLean M, Keefe DL, Liu L. Telomere susceptibility to cigarette smoke-induced oxidative damage and chromosomal instability of mouse embryos in vitro. Free Radic. Biol. Med. 2010;48:1663–1676. doi: 10.1016/j.freeradbiomed.2010.03.026. [DOI] [PubMed] [Google Scholar]
  • 155.Liu J, et al. Delay in oocyte aging in mice by the antioxidant N-acetyl-L-cysteine (NAC) Hum. Reprod. 2012;27:1411–1420. doi: 10.1093/humrep/des019. [DOI] [PubMed] [Google Scholar]
  • 156.Chen ZG, et al. Effects of plant polyphenols on ovarian follicular reserve in aging rats. Biochem. Cell Biol. 2010;88:737–745. doi: 10.1139/O10-012. [DOI] [PubMed] [Google Scholar]
  • 157.Tarín JJ, Pérez-Albalá S, Cano A. Oral antioxidants counteract the negative effects of female aging on oocyte quantity and quality in the mouse. Mol. Reprod. Dev. 2002;61:385–397. doi: 10.1002/mrd.10041. [DOI] [PubMed] [Google Scholar]
  • 158.Tarín JJ, Pérez-Albalá S, Pertusa JF, Cano A. Oral administration of pharmacological doses of Vitamins C and E reduces reproductive fitness and impairs the ovarian and uterine functions of female mice. Theriogenology. 2002;57:1539–1550. doi: 10.1016/S0093-691X(02)00636-2. [DOI] [PubMed] [Google Scholar]
  • 159.Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C. Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database Syst. Rev. 2012;2012:CD007176. doi: 10.1002/14651858.CD007176.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Ristow M, et al. Antioxidants prevent health-promoting effects of physical exercise in humans. Proc. Natl. Acad. Sci. USA. 2009;106:8665–8670. doi: 10.1073/pnas.0903485106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Evans JR, Lawrenson JG. Antioxidant vitamin and mineral supplements for slowing the progression of age-related macular degeneration. Cochrane Database Syst. Rev. 2017;7:CD000254. doi: 10.1002/14651858.CD000254.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Mathew MC, Ervin AM, Tao J, Davis RM. Antioxidant vitamin supplementation for preventing and slowing the progression of age-related cataract. Cochrane Database Syst. Rev. 2012;6:CD004567. doi: 10.1002/14651858.CD004567.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Rutjes AWS, et al. Vitamin and mineral supplementation for maintaining cognitive function in cognitively healthy people in mid and late life. Cochrane Database Syst. Rev. 2018;12:CD011906. doi: 10.1002/14651858.CD011906.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Al-Khudairy L, et al. Vitamin C supplementation for the primary prevention of cardiovascular disease. Cochrane Database Syst. Rev. 2017;3:CD011114. doi: 10.1002/14651858.CD011114.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Agarwal A, Durairajanayagam D, du Plessis SS. Utility of antioxidants during assisted reproductive techniques: an evidence based review. Reprod. Biol. Endocrinol. 2014;12:112–112. doi: 10.1186/1477-7827-12-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Tesarik J. Towards personalized antioxidant use in female infertility: need for more molecular and clinical studies. Biomedicines. 2021;9:1933. doi: 10.3390/biomedicines9121933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Ochiai A, et al. Influence of resveratrol supplementation on IVF-embryo transfer cycle outcomes. Reprod. Biomed. Online. 2019;39:205–210. doi: 10.1016/j.rbmo.2019.03.205. [DOI] [PubMed] [Google Scholar]
  • 168.Zhang LX, et al. Resveratrol (RV): a pharmacological review and call for further research. Biomed. Pharmacother. 2021;143:112164. doi: 10.1016/j.biopha.2021.112164. [DOI] [PubMed] [Google Scholar]
  • 169.Heilbronn LK, Ravussin E. Calorie restriction and aging: review of the literature and implications for studies in humans. Am. J. Clin. Nutr. 2003;78:361–369. doi: 10.1093/ajcn/78.3.361. [DOI] [PubMed] [Google Scholar]
  • 170.Féry F, Plat L, Balasse EO. Effects of metformin on the pathways of glucose utilization after oral glucose in non-insulin-dependent diabetes mellitus patients. Metabolism. 1997;46:227–233. doi: 10.1016/S0026-0495(97)90307-3. [DOI] [PubMed] [Google Scholar]
  • 171.Hundal RS, et al. Mechanism by which metformin reduces glucose production in type 2 diabetes. Diabetes. 2000;49:2063–2069. doi: 10.2337/diabetes.49.12.2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Onken B, Driscoll M. Metformin induces a dietary restriction–like state and the oxidative stress response to extend C. elegans healthspan via AMPK, LKB1, and SKN-1. PLoS One. 2010;5:e8758. doi: 10.1371/journal.pone.0008758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Martin-Montalvo A, et al. Metformin improves healthspan and lifespan in mice. Nat. Commun. 2013;4:2192. doi: 10.1038/ncomms3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Campbell JM, Bellman SM, Stephenson MD, Lisy K. Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: a systematic review and meta-analysis. Ageing Res. Rev. 2017;40:31–44. doi: 10.1016/j.arr.2017.08.003. [DOI] [PubMed] [Google Scholar]
  • 175.Isola JVV, et al. Mild calorie restriction, but not 17α-estradiol, extends ovarian reserve and fertility in female mice. Exp. Gerontol. 2022;159:111669. doi: 10.1016/j.exger.2021.111669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Selesniemi K, Lee H-J, Tilly JL. Moderate caloric restriction initiated in rodents during adulthood sustains function of the female reproductive axis into advanced chronological age. Aging Cell. 2008;7:622–629. doi: 10.1111/j.1474-9726.2008.00409.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Qin X, et al. Metformin prevents murine ovarian aging. Aging. 2019;11:3785–3794. doi: 10.18632/aging.102016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Xu S, et al. Glucose activates the primordial follicle through the AMPK/mTOR signaling pathway. Clin. Transl. Med. 2020;10:e122. doi: 10.1002/ctm2.122. [DOI] [Google Scholar]
  • 179.Oner G, Ozcelik B, Ozgun MT, Ozturk F. The effects of metformin and letrozole on endometrium and ovary in a rat model. Gynecol. Endocrinol. 2011;27:1084–1086. doi: 10.3109/09513590.2011.589928. [DOI] [PubMed] [Google Scholar]
  • 180.Barilovits SJ, et al. Characterization of a mechanism to inhibit ovarian follicle activation. Fertil. Steril. 2014;101:1450–1457. doi: 10.1016/j.fertnstert.2014.01.025. [DOI] [PubMed] [Google Scholar]
  • 181.Lane MA, Ingram DK, Roth GS. 2-Deoxy-D-glucose feeding in rats mimics physiologic effects of calorie restriction. J. Anti Aging Med. 1998;1:327–337. doi: 10.1089/rej.1.1998.1.327. [DOI] [Google Scholar]
  • 182.Minor RK, et al. Chronic ingestion of 2-deoxy-D-glucose induces cardiac vacuolization and increases mortality in rats. Toxicol. Appl. Pharmacol. 2010;243:332–339. doi: 10.1016/j.taap.2009.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Chiang JL, et al. Mitochondria in ovarian aging and reproductive longevity. Ageing Res. Rev. 2020;63:101168. doi: 10.1016/j.arr.2020.101168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Wang, T., Zhang, M., Jiang, Z., Seli, E. Mitochondrial dysfunction and ovarian aging. Am. J. Reprod. Immunol.77, bqaa001 (2017). [DOI] [PubMed]
  • 185.Camacho-Pereira J, et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab. 2016;23:1127–1139. doi: 10.1016/j.cmet.2016.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Yang Q, et al. Increasing ovarian NAD(+) levels improve mitochondrial functions and reverse ovarian aging. Free Radic. Biol. Med. 2020;156:1–10. doi: 10.1016/j.freeradbiomed.2020.05.003. [DOI] [PubMed] [Google Scholar]
  • 187.Bertoldo MJ, et al. NAD+ repletion rescues female fertility during reproductive aging. Cell Rep. 2020;30:1670–1681.e7. doi: 10.1016/j.celrep.2020.01.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Yang L, et al. Mitochondrial DNA mutation exacerbates female reproductive aging via impairment of the NADH/NAD(+) redox. Aging Cell. 2020;19:e13206. doi: 10.1111/acel.13206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Bonkowski MS, Sinclair DA. Slowing ageing by design: the rise of NAD(+) and sirtuin-activating compounds. Nat. Rev. Mol. Cell Biol. 2016;17:679–690. doi: 10.1038/nrm.2016.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Jia K. The TOR pathway interacts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span. Development. 2004;131:3897–3906. doi: 10.1242/dev.01255. [DOI] [PubMed] [Google Scholar]
  • 191.Vellai T, et al. Genetics influence of TOR kinase on lifespan in C. elegans. Nature. 2003;426:620–620. doi: 10.1038/426620a. [DOI] [PubMed] [Google Scholar]
  • 192.Kapahi P, et al. Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr. Biol. 2004;14:885–890. doi: 10.1016/j.cub.2004.03.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Lamming DW, et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science. 2012;335:1638–1643. doi: 10.1126/science.1215135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Wu JJ, et al. Increased mammalian lifespan and a segmental and tissue-specific slowing of aging after genetic reduction of mTOR expression. Cell Rep. 2013;4:913–920. doi: 10.1016/j.celrep.2013.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Miller RA, et al. Rapamycin-mediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction. Aging Cell. 2014;13:468–477. doi: 10.1111/acel.12194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Neff F, et al. Rapamycin extends murine lifespan but has limited effects on aging. J. Clin. Invest. 2013;123:3272–3291. doi: 10.1172/JCI67674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Chen C, Liu Y, Liu Y, Zheng P. mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci. Signal. 2009;2:ra75. doi: 10.1126/scisignal.2000559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Miller RA, et al. Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. J. Gerontol. A Biol. Sci. Med. Sci. 2011;66:191–201. doi: 10.1093/gerona/glq178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Harrison DE, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395. doi: 10.1038/nature08221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Guo Z, Yu Q. Role of mTOR signaling in female reproduction. Front. Endocrinol. 2019;10:692–692. doi: 10.3389/fendo.2019.00692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Adhikari D, et al. Tsc/mTORC1 signaling in oocytes governs the quiescence and activation of primordial follicles. Hum. Mol. Genet. 2010;19:397–410. doi: 10.1093/hmg/ddp483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Adhikari D, et al. Disruption of Tsc2 in oocytes leads to overactivation of the entire pool of primordial follicles. Mol. Hum. Reprod. 2009;15:765–770. doi: 10.1093/molehr/gap092. [DOI] [PubMed] [Google Scholar]
  • 203.Zhang H, et al. Somatic cells initiate primordial follicle activation and govern the development of dormant oocytes in mice. Curr. Biol. 2014;24:2501–2508. doi: 10.1016/j.cub.2014.09.023. [DOI] [PubMed] [Google Scholar]
  • 204.Lu X, et al. Stimulation of ovarian follicle growth after AMPK inhibition. Reproduction. 2017;153:683–694. doi: 10.1530/REP-16-0577. [DOI] [PubMed] [Google Scholar]
  • 205.Tanaka Y, et al. Deletion of tuberous sclerosis 1 in somatic cells of the murine reproductive tract causes female infertility. Endocrinology. 2012;153:404–416. doi: 10.1210/en.2011-1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Choi J, Jo M, Lee E, Choi D. AKT is involved in granulosa cell autophagy regulation via mTOR signaling during rat follicular development and atresia. Reproduction. 2014;147:73–80. doi: 10.1530/REP-13-0386. [DOI] [PubMed] [Google Scholar]
  • 207.Zhang XM, et al. Rapamycin preserves the follicle pool reserve and prolongs the ovarian lifespan of female rats via modulating mTOR activation and sirtuin expression. Gene. 2013;523:82–87. doi: 10.1016/j.gene.2013.03.039. [DOI] [PubMed] [Google Scholar]
  • 208.Tong Y, et al. Rapamycin-sensitive mTORC1 signaling is involved in physiological primordial follicle activation in mouse ovary. Mol. Reprod. Dev. 2013;80:1018–1034. doi: 10.1002/mrd.22267. [DOI] [PubMed] [Google Scholar]
  • 209.Adhikari D, et al. Pharmacological inhibition of mTORC1 prevents over-activation of the primordial follicle pool in response to elevated PI3K signaling. PLoS One. 2013;8:e53810. doi: 10.1371/journal.pone.0053810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Luo LL, Xu JJ, Fu YC. Rapamycin prolongs female reproductive lifespan. Cell Cycle. 2013;12:3353–3354. doi: 10.4161/cc.26578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Bitto A, et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. Elife. 2016;5:e16351. doi: 10.7554/eLife.16351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Wang Y, et al. The essential role of PRAK in tumor metastasis and its therapeutic potential. Nat. Commun. 2021;12:1736. doi: 10.1038/s41467-021-21993-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Woo Y, et al. Rapamycin promotes ROS-mediated cell death via functional inhibition of xCT expression in melanoma under gamma-irradiation. Front. Oncol. 2021;11:665420. doi: 10.3389/fonc.2021.665420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Orentreich N, Brind JL, Rizer RL, Vogelman JH. Age changes and sex differences in serum dehydroepiandrosterone sulfate concentrations throughout adulthood. J. Clin. Endocrinol. Metab. 1984;59:551–555. doi: 10.1210/jcem-59-3-551. [DOI] [PubMed] [Google Scholar]
  • 215.Zhang J, et al. Dehydroepiandrosterone improves the ovarian reserve of women with diminished ovarian reserve and is a potential regulator of the immune response in the ovaries. BioScience Trends. 2015;9:350–359. doi: 10.5582/bst.2015.01154. [DOI] [PubMed] [Google Scholar]
  • 216.Gleicher N, Weghofer A, Barad DH. Improvement in diminished ovarian reserve after dehydroepiandrosterone supplementation. Reprod. Biomed. Online. 2010;21:360–365. doi: 10.1016/j.rbmo.2010.04.006. [DOI] [PubMed] [Google Scholar]
  • 217.Narkwichean A, et al. Effects of dehydroepiandrosterone on in vivo ovine follicular development. Hum. Reprod. 2013;29:146–154. doi: 10.1093/humrep/det408. [DOI] [PubMed] [Google Scholar]
  • 218.Meredith S, Jackson K, Dudenhoeffer G, Graham L, Epple J. Long-term supplementation with melatonin delays reproductive senescence in rats, without an effect on number of primordial follicles. Exp. Gerontol. 2000;35:343–352. doi: 10.1016/S0531-5565(00)00092-9. [DOI] [PubMed] [Google Scholar]
  • 219.Fernández BE, Díaz E, Fernández C, Núñez P, Díaz B. Ovarian aging: melatonin regulation of the cytometric and endocrine evolutive pattern. Curr. Aging Sci. 2013;6:1–7. doi: 10.2174/1874609811306010001. [DOI] [PubMed] [Google Scholar]
  • 220.Tamura, H. et al. Long-term melatonin treatment delays ovarian aging. J. Pineal. Res.62, (2017). [DOI] [PubMed]
  • 221.Song C, et al. Melatonin improves age-induced fertility decline and attenuates ovarian mitochondrial oxidative stress in mice. Sci. Rep. 2016;6:35165. doi: 10.1038/srep35165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Zhang L, et al. Melatonin regulates the activities of ovary and delays the fertility decline in female animals via MT1/AMPK pathway. J. Pineal. Res. 2019;66:e12550. doi: 10.1111/jpi.12550. [DOI] [PubMed] [Google Scholar]
  • 223.Yang C, et al. Melatonin delays ovarian aging in mice by slowing down the exhaustion of ovarian reserve. Commun. Biol. 2021;4:534. doi: 10.1038/s42003-021-02042-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Bellipanni G, Bianchi P, Pierpaoli W, Bulian D, Ilyia E. Effects of melatonin in perimenopausal and menopausal women: a randomized and placebo controlled study. Exp. Gerontol. 2001;36:297–310. doi: 10.1016/S0531-5565(00)00217-5. [DOI] [PubMed] [Google Scholar]
  • 225.Takasaki A, Nakamura Y, Tamura H, Shimamura K, Morioka H. Melatonin as a new drug for improving oocyte quality. Reprod. Med. Biol. 2003;2:139–144. doi: 10.1111/j.1447-0578.2003.00035.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Tamura H, et al. Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rate. J. Pineal. Res. 2008;44:280–287. doi: 10.1111/j.1600-079X.2007.00524.x. [DOI] [PubMed] [Google Scholar]
  • 227.Batioglu AS, Sahin U, Gurlek B, Ozturk N, Unsal E. The efficacy of melatonin administration on oocyte quality. Gynecol. Endocrinol. 2012;28:91–93. doi: 10.3109/09513590.2011.589925. [DOI] [PubMed] [Google Scholar]
  • 228.Fernando S, et al. Melatonin in assisted reproductive technology: a pilot double-blind randomized placebo-controlled clinical trial. Front. Endocrinol. 2018;9:545. doi: 10.3389/fendo.2018.00545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Espino J, et al. Impact of melatonin supplementation in women with unexplained infertility undergoing fertility treatment. Antioxidants. 2019;8:338. doi: 10.3390/antiox8090338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.George K, Kamath MS. Fertility and age. J. Hum. Reprod. Sci. 2010;3:121–123. doi: 10.4103/0974-1208.74152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Smith KR, et al. Familial aggregation of survival and late female reproduction. J. Gerontol. A Biol. Sci. Med. Sci. 2009;64:740–744. doi: 10.1093/gerona/glp055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Abdollahifar MA, et al. Vitamin C restores ovarian follicular reservation in a mouse model of aging. Anat. Cell Biol. 2019;52:196–203. doi: 10.5115/acb.2019.52.2.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Zhuang XL, et al. Effects of genistein on ovarian follicular development and ovarian life span in rats. Fitoterapia. 2010;81:998–1002. doi: 10.1016/j.fitote.2010.06.018. [DOI] [PubMed] [Google Scholar]
  • 234.Appt SE, et al. The effect of diet and cardiovascular risk on ovarian aging in cynomolgus monkeys (Macaca fascicularis) Menopause. 2010;17:741–748. doi: 10.1097/gme.0b013e3181d20cd2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Liu M, et al. Resveratrol protects against age-associated infertility in mice. Hum. Reprod. 2013;28:707–717. doi: 10.1093/humrep/des437. [DOI] [PubMed] [Google Scholar]
  • 236.Ben-Meir A, et al. Coenzyme Q10 restores oocyte mitochondrial function and fertility during reproductive aging. Aging Cell. 2015;14:887–895. doi: 10.1111/acel.12368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Timoteo-Ferreira F, et al. Apocynin dietary supplementation delays mouse ovarian ageing. Oxid. Med. Cell Longev. 2019;2019:5316984. doi: 10.1155/2019/5316984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Silva E, et al. Antioxidant supplementation modulates age-related placental bed morphology and reproductive outcome in mice. Biol. Reprod. 2015;93:56. doi: 10.1095/biolreprod.114.127746. [DOI] [PubMed] [Google Scholar]
  • 239.Xian Y, et al. Antioxidants retard the ageing of mouse oocytes. Mol. Med. Rep. 2018;18:1981–1986. doi: 10.3892/mmr.2018.9167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Akino N, et al. Activation of Nrf2/Keap1 pathway by oral Dimethylfumarate administration alleviates oxidative stress and age-associated infertility might be delayed in the mouse ovary. Reprod. Biol. Endocrinol. 2019;17:23. doi: 10.1186/s12958-019-0466-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Wei M, et al. Ovarian failure-resistant effects of catalpol in aged female rats. Biol. Pharm. Bull. 2014;37:1444–1449. doi: 10.1248/bpb.b14-00064. [DOI] [PubMed] [Google Scholar]
  • 242.Jinno M, et al. Low-dose metformin improves pregnancy rate in IVF repeaters without polycystic ovary syndrome: its indication and mechanism. Fertil. Steril. 2010;94:S29–S29. doi: 10.1016/j.fertnstert.2010.07.110. [DOI] [Google Scholar]
  • 243.Zhang Z, et al. alpha-ketoglutarate delays age-related fertility decline in mammals. Aging Cell. 2021;20:e13291. doi: 10.1111/acel.13291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Yu J, Yaba A, Kasiman C, Thomson T, Johnson J. mTOR controls ovarian follicle growth by regulating granulosa cell proliferation. PLoS One. 2011;6:e21415. doi: 10.1371/journal.pone.0021415. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cell Research are provided here courtesy of Nature Publishing Group

RESOURCES