5,084
Views
0
CrossRef citations to date
0
Altmetric
Review

The influence of the gut microbiome on ovarian aging

ORCID Icon, , , ORCID Icon, , ORCID Icon & ORCID Icon show all
Article: 2295394 | Received 04 Jul 2023, Accepted 12 Dec 2023, Published online: 03 Jan 2024

ABSTRACT

Ovarian aging occurs prior to the aging of other organ systems and acts as the pacemaker of the aging process of multiple organs. As life expectancy has increased, preventing ovarian aging has become an essential goal for promoting extended reproductive function and improving bone and genitourinary conditions related to ovarian aging in women. An improved understanding of ovarian aging may ultimately provide tools for the prediction and mitigation of this process. Recent studies have suggested a connection between ovarian aging and the gut microbiota, and alterations in the composition and functional profile of the gut microbiota have profound consequences on ovarian function. The interaction between the gut microbiota and the ovaries is bidirectional. In this review, we examine current knowledge on ovary-gut microbiota crosstalk and further discuss the potential role of gut microbiota in anti-aging interventions. Microbiota-based manipulation is an appealing approach that may offer new therapeutic strategies to delay or reverse ovarian aging.

Introduction

The gut microbiota, which is the second genome of the human body, plays an important role in human health and pathogenesis. The gut microbiota is a complex symbiotic ecosystem, and imbalances in the gut microbiota have been widely reported in a range of common chronic diseases, such as type 2 diabetes, cardiometabolic diseases and metabolic liver diseases.Citation1 Mounting evidence has confirmed that there is a bidirectional relationship between the gut and numerous crucial organs of the host, in which the microbiota serves as a regulator.Citation2,Citation3

Significant changes in the identities of dominant microorganisms, bacterial diversity, and functional features have been uncovered in human aging and age-related diseases.Citation4,Citation5 A recent study showed the existence of multiple “clocks” within the body: organs/systems age at different rates, and the aging rate of specific organs or systems correlates with that of the gut microbiome.Citation6 Consequently, individuals may present with different health or disease states. Gut microbial features can be used to predict healthy aging or mortality in elderly individuals.Citation7,Citation8 Interestingly, experiments on mice demonstrated that transplantation of aged donor microbiota accelerates specific processes of age-associated brain degeneration in the recipient, and conversely, transplantation of young donor microbiota can reverse this condition.Citation9,Citation10 It has been suggested that the gut microbiota could serve as a potential therapeutic target for new anti-aging interventions.Citation5

Ovarian aging refers to the gradual decline in ovarian function with age. It is characterized by the gradual deterioration in oocyte quantity and quality, accompanied by menstrual cycle irregularity, infertility and finally, the cessation of menses.Citation11 The ovary experiences a different lifespan from other organs. Recently, multiomics profiling identified a significant acceleration in female biological aging around the third and fifth decades of life, which coincided with the time points of fertility decline and menopausal status.Citation12 Thus, ovarian aging is considered the pacemaker of aging in the female body, driving the aging of multiple organs.Citation13 The levels of sex hormones, including follicle-stimulating hormone (FSH), luteinizing hormone (LH) and anti-Müllerian hormone (AMH), were highly correlated with several other aging clocks.Citation12 As life expectancy increases worldwide, ovarian aging will gradually become a major health problem for women.Citation14 Ovarian aging is an extremely complex process with causes that have not yet been fully clarified. Growing evidence shows that there is a link between the gut microbiota and ovarian function,Citation15 and the gut microbiota-ovary axis contributes to follicular development.Citation16 In this review, we aim to synthesize different aspects of ovary-microbiota crosstalk and provide meaningful conclusions (). We also try to explain the potential role of the microbiota in some anti-aging approaches.

Figure 1. The physiologic succession of gut microbiota across natural ovarian aging.

Figure 1. The physiologic succession of gut microbiota across natural ovarian aging.

The gut microbiota and natural ovarian aging

Menopause is the hallmark event of natural ovarian aging. Natural menopause, which mostly occurs between the ages of 49 and 52 years, is diagnosed retrospectively after a 12-month cessation of spontaneous menses without pathological causes.Citation17 As the finite stores of ovarian follicles are depleted and the remaining follicles lose their sensitivity to gonadotropins, the follicles stop developing and secreting the ovarian hormones estrogen and progesterone.Citation18

Estradiol (E2) levels only decrease relatively late during the process of perimenopausal transition.Citation11 However, a dramatic decline in plasma E2 levels occurs during the final menstrual cycle, and afterward, the postmenopausal ovaries stop synthesizing E2.Citation19 It is known that the gut microbiome is one of the important regulators of circulating estrogens.Citation20 Gut microbial β-glucuronidase (gmGUS) can transform estrogen from deactivated forms to active forms and further affect estrogen levels in the host.Citation21,Citation22 Gut microbiota dysbiosis and lower microbial diversity result in a reduction in gmGUS and alterations in systemic estrogens.Citation15 Gut bacteria, in turn, are also influenced by estrogen. It has been reported that the relative abundances of two bacterial taxa, namely, the Gammaproteobacteria class and an unknown family from Mixococcales, were positively correlated with E2 levels, and the abundance of the family Prevotellaceae was negatively correlated with E2 levels.Citation23,Citation24 Furthermore, estrogen or estrogen-like compounds exert microbiome-modulating effects and maintain gut epithelial barrier integrity in mice with metabolic syndrome.Citation25 Supplementation with estrogen-like foods such as soy isoflavones augments the abundances of the genera Bifidobacterium and Eubacterium while restraining the genera Lactobacillus and unclassified Clostridiaceae in postmenopausal women.Citation26 Under certain pathological conditions, dysbiosis of the gut microbiota and elevated gmGUS activity may contribute to the development of estrogen-driven diseases and menopause-related disorders by participating in the circulation or activation of endogenous and exogenous estrogen-like compounds.Citation21

Menopausal status is known to affect the gut microbiota. Differences in the composition and the functional profile of the gut microbiota between women of different menopausal statuses have already been reported as described below. A total of 90 differentially abundant taxa between premenopausal (n = 44) and postmenopausal women (n = 45) were observed, and the pyrimidine and one-carbon pools associated with the folate pathways were enriched in premenopausal women compared with postmenopausal women.Citation27 The genus Roseburia was identified as the most remarkable bacterial taxon to discriminate between premenopausal (n = 17) and postmenopausal groups (n = 20), and higher propanoate and butanoate metabolism in premenopausal women was observed.Citation23 In contrast, Zhao et al. demonstrated that the alpha diversity of microbiomes in postmenopausal women (n = 24) was significantly lower than that in premenopausal women (n = 24), the abundances of the phylum Firmicutes and the genus Roseburia were low, the abundances of the phylum Bacteroidetes and the toluene-producing genus Tolumonas were enriched in postmenopausal women, and homocysteine synthesis-related processes were activated in postmenopausal women.Citation28 Other microbes, such as the genera A. odoratum and B. cholerae, were also enriched in postmenopausal women (n = 90) compared with premenopausal women (n = 66).Citation24 Remarkably, Firmicutes and Bacteroidetes are dominant groups of beneficial bacteria in the human gutCitation29 and Roseburia is a butyrate-producing bacteria.Citation30,Citation31 Although taxa across studies varied, it might be attributed to the different study populations, sample sizes or methods.

In an animal model, alterations in the gut microbiota induced by ovarian aging were also described. 4-Vinylcyclohexene diepoxide (VCD), an occupational chemical that directly kills primordial and primary ovarian follicles of the ovary but shows no toxicity toward extraovarian tissue, induces ovarian aging analogous to natural ovarian aging.Citation32 Researchers determined that there were significant differences in both the alpha and beta diversity between the mice in the VCD group and the control group.Citation33 The levels of several genera, including Helicobacter, Odoribacter, and Alistipes, were lower, and Clostridium XIVa, Barnesiella, Bacteroides, and Mucispirillum were higher among the dominant microbiota of the VCD group.Citation33 Studies in Drosophila revealed that the intestinal epithelium deteriorates as females age, which leads to high microbiota loads, barrier impairment, and decreased lifespan.Citation34,Citation35 The steroid hormone ecdysone, produced by active ovaries of the fly, could stimulate the division and expansion of intestinal stem cells and enhance their reproductive output.Citation36

To further probe the more complicated and deeper causal linkage between the gut microbiota and natural ovarian aging, fecal microbiota transplantation (FMT) was applied to naturally aging mice. Aged mice (42 weeks old) treated with FMT from young donors (5 weeks old) presented a “young animal-like phenotype” in the gut microbiome, and follicular atresia and apoptosis were reduced to improve the fertility of aged mice.Citation37 These ovarian aging-delay effects might be explained by improving the inflammatory status and immune microenvironment.Citation37,Citation38 Substantial evidence in aged zebrafish also demonstrated the effect of the young microbiota on reproductive modulation. FMT from young donors to aged zebrafish could counteract the adverse effects of subsequent exposure to toxic pollutants, potently reduce the levels of membrane-bound progestin receptor transcripts, promote the progression of oogenesis, and reduce the rate of malformations caused by toxic pollutants, and their offspring presented a stronger locomotor capability and swam much faster.Citation39 Overall, maintenance of a “youthful” gut microbiota during ovarian aging may limit ovarian senescence and provide additional benefits.

The gut microbiota and premature ovarian insufficiency

Rather than natural ovarian aging, some women in the population experience a premature decline in ovarian function, that is, premature ovarian insufficiency (POI).Citation40 POI is a clinical syndrome defined by exhaustion of normal ovarian activity in patients younger than 40 years, which presents as a menstrual disorder (amenorrhea or oligomenorrhea) with elevated gonadotropin levels and low E2 levels.Citation41 The estimated global prevalence is as high as 3.7%, while 0.1% of women aged less than 30 and 0.01% of women aged less than 20 present with the condition.Citation42 POI has attracted public attention because of its adverse effects on fertility and subsequent negative health impacts, such as increased risk of cardiovascular diseases, cognitive impairment, mood and sexual dysfunction, osteoporosis and shorter life expectancy.Citation43–45 POI is a highly heterogeneous condition, with 70 to 90% of POI cases having unknown causes in addition to known genetic, autoimmune, infectious, and iatrogenic factors, that is idiopathic POI.Citation42 Recently, mounting evidence has suggested that gut microbiota dysbiosis is associated with POI. In this section, we review the current research linking gut microbiota and POI, focusing on idiopathic POI, iatrogenic POI and POI-driven diseases.

Idiopathic POI

It has been noted that there was a reduction in beta diversity in patients with POI.Citation46 At the phylum level, higher Bacteroides and lower Firmicutes abundances were observed in POI patients, while higher Butyricimonas, Dorea, Lachnobacterium, and Sutterella and lower Bulleidia and Faecalibacterium abundances at the genus level were observed in women with POI (n = 35) than in healthy women (n = 18).Citation47 These microbial changes were correlated with the levels of FSH, LH, E2, and AMH and the FSH/LH ratio.Citation47 In addition, decreases in the abundances of the genera Eggerthella and Staphylococcus and increases in the abundances of the genera Comamonas and Barnesiella were observed in POI patients (n = 10) relative to controls (n = 10).Citation46 Researchers have also revealed positive associations between Eggerthella and several serum metabolites.Citation46 Although POI shares similar clinical manifestations with natural ovarian aging, few studies have reported differences in the gut microbiota between them.

Hormone replacement therapy (HRT) is the primary recommended treatment given to women with POI both for symptom management and disease prevention.Citation40 Jiang et al. determined that the alterations in beta diversity, relative abundance of the genus Eggerthella, serum metabolites, and E2 and progesterone hormone levels related to POI were significantly reversed after HRT implementation,Citation46 which further validated the interaction between estrogen and gut microbiota. Animal experiments confirmed that E. lenta gavage induced ovarian fibrosis in mice and that estrogen treatment alleviated ovarian fibrosis.Citation46 Despite the potential linkage between POI and the gut microbiota, the causal nature of this relationship still needs to be deeply investigated.

Taken together, these results show that gut microbiota dysbiosis, such as disturbance of core microbiota (Lachnoclostridium and Bacteroides), short-chain fatty acid (SCFA) producers (Bacteroides and Faecalibacterium) and an increase in opportunistic pathogenic bacteria (Eggerthella), may be involved in the pathology of POI, and the reversal of gut microbiota may be a new strategy for ovary preservation to some extent.

Iatrogenic POI

As noted above, most POI cases are idiopathic. However, among the few cases with known causes, iatrogenic factors account for a large proportion.Citation48–50 Many medical factors can damage ovarian function, such as pelvic radiotherapy, chemotherapy and surgery, in tumor patients, which can lead to premature decline or even total loss of ovarian function.Citation51 Specifically, the higher the therapeutic dose, the larger the radiation area and the larger the surgical area, the greater the impact on ovarian function.Citation52–55 Patients who have undergone ovarian surgery, especially laparoscopic surgery, are candidates for iatrogenic POI.Citation48 Bone loss and genitourinary atrophy are two major challenges faced by many young cancer patients and postsurgical women due to iatrogenic ovarian injury, which seriously impacts their overall quality of life.Citation50

A study profiled and analyzed the changes in gut microbiota in ovarian cancer patients who underwent different anticancer treatments and found that postsurgery samples exhibited a significant decrease in the abundances of the phyla Bacteroidetes and Firmicutes and a significant increase in the abundance of the phylum Proteobacteria compared with presurgery samples; however, after-chemotherapy samples exhibited significantly increased abundances of the phyla Bacteroidetes and Firmicutes and a decreased abundance of the phylum Proteobacteria compared with before.Citation56

Bilateral ovariectomy is the most mature method to build an iatrogenic menopause animal model and has been widely used in menopause and menopause-related research, such as menopausal osteoporosis and genitourinary atrophy.Citation53–59 Therefore, we focused on mouse ovariectomy studies to demonstrate the relationship and mechanism of the gut microbiome and iatrogenic menopause. Animal studies have shown that changes in the gut microbiota are associated with iatrogenic ovarian aging and associated relevant adverse outcomes.

Many studies based on an ovariectomized (OVX) mouse model have revealed that the gut microbiota could be affected by iatrogenic menopause.Citation60 Significant differences in beta diversityCitation61–64 and alpha diversityCitation62 have been observed between the gut microbiota of the OVX group and the sham group. Several studies found that the OVX group had a higher Firmicutes/Bacteroidetes ratio and lower relative abundance of Deferribacteres at the phylum level.Citation65–68 At the genus level, the abundance of the predominant genera in the sham group decreased significantly in the OVX group (87% vs. 29%).Citation62 In contrast, lower-abundance genera in the sham group were importantly enriched in the OVX group (<5% vs. 63%).Citation62 Zhang et al. identified four key significant genera in OVX rats: Incertae_Sedis, Anaerovorax, Anaerotruncus and Helicobacter.Citation63 Furthermore, the OVX group exhibited apparent weight gain,Citation65 intestinal barrier impairmentCitation61, bone lossCitation65,Citation66 and vaginal atrophy.Citation64 The gut microbiota is indispensable for OVX-induced osteoclast differentiation, and germ-free (GF) animals were protected against osteoporosis induced by OVX. The Firmicutes Bacteroidetes phylum and the Firmicutes/Bacteroidetes ratio were critical in osteoclast differentiation, and supplementation with the probiotic Lactobacillus salivarius LI01 from the Firmicutes phylum prevented OVX-induced osteoporosis in mice.Citation67 FMT from ovary-intact mice inhibited the gut microbiota changes and weight gain induced by OVX, suppressed the release of pro-osteoclastogenic cytokines, increased fecal SCFA levels (mainly acetic acid and propionic acid), reduced intestinal permeability and ultimately prevented OVX-induced bone loss.Citation65 Surprisingly, Huang et al. showed that FMT from ovary-intact fecund females changed the gut microbiota and significantly alleviated vaginal epithelial atrophy in OVX mice.Citation64 In general, FMT might act as a promising target for the prevention and treatment of iatrogenic ovarian aging-related consequences ().

Figure 2. FMT alter the gut microbiota and slow the progression of ovarian aging related diseases.

Figure 2. FMT alter the gut microbiota and slow the progression of ovarian aging related diseases.

Association among antiaging interventions, ovarian function and intestinal microbes

Antiaging drugs

To date, no clinically feasible techniques are available to either delay or reverse ovarian dysfunction associated with advanced age.Citation69 However, important advances have been made in the field of anti-ovarian aging during the past decades, and numerous emerging antiaging agents, such as coenzyme Q10 (CoQ10), melatonin, nicotinamide mononucleotide (NMN), resveratrol, rapamycin and N-acetyl-L-cysteine (NAC), have shown great potential to delay or reverse ovarian aging.Citation68–70 Although no study has directly explored the relationship among these antiaging drugs, ovarian function and gut microbiota, some indirect evidence has shown that the beneficial effects of these drugs may be associated with gut microbes. As shown in , these drugs may work through intestinal microbes.Citation102

Table 1. Association among antiaging drugs, ovarian function and intestinal microbes.

CoQ10

CoQ10 is a fat-soluble, vitamin-like benzoquinone compound, plays a critical role in cellular energy production and functions as an antioxidant in its reduced form.Citation69 The synthesis of CoQ10 appears to decrease with age, which coincided with the time point of fertility and oocyte quality decline.Citation103 Pretreatment with CoQ10 during the fertility treatment improved ovarian response and the quality of embryo in young patient with diminished ovarian reserve.Citation104 CoQ10 treatment in mice not only preserved the ovarian follicle pool but also promoted ovulation of gametes capable of supporting normal development.Citation68,Citation103 Dietary CoQ10 supplementation also had beneficial effects on the reproductive variables of aged hensCitation105 gestating sows with high parityCitation91 and red tilapia.Citation82 However, in women with infertility undergoing assisted reproductive technology procedures, CoQ10 increased clinical pregnancy but showed no effect on live birth or miscarriage rates.Citation106 CoQ10 also has an impact on the gut microbiota and the subsequent biomarkers it produces. Three-week of administration of CoQ10 resulted in 2.4 times and 7.5 times increase in the relative abundance of Ruminococcus and Lachnospiraceae AC 2044 groups, respectively, as well as a 1.26-fold increase in butyrate.Citation77 Pumpkin juice rich in CoQ10 gavage increased the abundance of Lactobacillus and Bifidobacterium and further protected the gut barrier by reducing Proteobacteria of mice.Citation72

Melatonin

Melatonin is an indoleamine produced by all cells, especially those in the pineal gland.Citation76 As a powerful antioxidant, melatonin has been shown to delay the decline in fertility in female animals.Citation71,Citation73 Oxidative stress (OS) is considered a key factor in ovarian aging that accelerates follicle loss and atresia,Citation74 along with significant gut microbiota dysbiosisCitation73,Citation107 while melatonin reverses OS-induced phenotypes and improves gut microbiota communities.Citation75 Melatonin treatments in mice not only significantly increased the number and quality of oocytes but also led to more blastocyst generation after in vitro fertilization and larger litter sizes than those in the controls.Citation73 Hao et al. identified that melatonin might increase the levels of E2 and LH as well as the levels of immunoglobulin and the numbers of growing follicles in aged laying hens.Citation71 Melatonin pretreatment reduced the Firmicutes abundance and the Firmicutes/Bacteroidetes ratio and increased the content of Bacteroidetes at the phylum level.Citation108 Additionally, oral melatonin supplementation significantly reversed gut microbiota dysbiosis induced by a high-fat diet (HFD), including the decreased alpha diversity of the intestinal microbiota, the decreased relative abundances of the genera Bacteroides and Alistipes, and the increased relative abundances of the order Lactobacillales and the genus Lactobacillus.Citation109 The marked correlation between acetic acid production and the relative abundances of the genera Bacteroides and Alistipes after FMT from melatonin-treated mice indicated that gut microbiota reprogramming might be the potential mechanism of melatonin treatment.Citation109

Nmn

NMN is another promising anti-ovarian aging drug. The loss of oocyte quality with reproductive aging accompanies declining levels of nicotinamide adenine dinucleotide (NAD+),Citation81 and as an intermediate of NAD+ biosynthesis, NAD+ can be compensated by NMN supplementation to play a role in delaying ovarian aging.Citation107 Twenty-week oral administration of NMN to middle-aged mice improved the estrous cycle condition, increased the number of follicles at different stages and the corpora lutea and serum FSH, and enhanced mitochondrial biogenesis, autophagy, and protease activity of granulosa cells (GCs) compared with the control treatment.Citation110 In HFD-induced POI mice, NMN administration reduced ovarian inflammation and the adipose size of abdominal fat tissue and improved oocyte quality.Citation70 NMN treatment increased the abundance of butyrate-producing bacterial genera, such as Ruminococcae_UCG-014, Prevotellaceae_NK3B31_group and the probiotic Akkermansia muciniphila, while decreasing the abundances of several harmful bacterial genera, such as Oscillibacter and Bilophila.Citation78 NMN also reduced intestinal mucosal impairment and maintained mucosal barrier integrity.Citation78

Resveratrol

Resveratrol, a natural polyphenol, potentially delays ovarian aging and protects against age-associated infertilityCitation79,Citation111,Citation112 and is capable of restoring the microbiota composition and reversing intestinal dysbiosis.Citation83 A study revealed that resveratrol ameliorated the oxidative stress-induced reduction in egg-laying rate and serum hormone level changes, as well as gut microbiota dysbiosis, and elevated the concentrations of the main SCFAs in laying hens.Citation84 This study directly proved that resveratrol modulated ovarian function through intestinal microbes. Enrichment of Bacteroides, Lachnospiraceae_NK4A136_group, Blautia, Lachnoclostridium, Parabacteroides and Ruminiclostridium_9, collectively referred to as the resveratrol-microbiota, displayed anti-obesity functions.Citation113

Rapamycin

The mammalian target of rapamycin (mTOR) signaling pathway exerts a vital role in folliculogenesis in female germline cells.Citation114 Activation of the AKT/mTOR signaling pathway in POI patients has been reported.Citation115 Suppressing mTOR signaling in HFD-induced POI mice reversed follicle loss and extended the ovarian lifespan.Citation116 Rapamycin is a natural macrolide compound initially segregated from bacteria and serves as a TOR signaling inhibitor.Citation85 Rapamycin treatment increased ovarian reserveCitation86 and prolonged ovarian lifespan with oocyte quality improvement, especially in aged mice.Citation87 In a cyclophosphamide-induced POI model, rapamycin prevented primordial follicle loss and protected the ovarian reserve.Citation88 A significant increase in the levels of segmented filamentous bacteria (Candidatus Arthromitus sp.) in rapamycin-treated mice was observed.Citation89 However, although rapamycin rescued intestinal barrier dysfunction and increased microbial loads in Drosophila, its antiaging effects seemed to be independent of the gut microbiota.Citation92

Nac

NAC is synthesized to help stimulate the synthesis of glutathione to exert antioxidant effects.Citation94 A study conducted in mice proposed that the administration of NAC could improve the quality of preimplantation oocytes and increased the total blastocyst cell number and litter sizes, thus delaying the fertility decline with reproductive aging.Citation96 NAC alleviated gut dysbiosis in HFD-fed mice by promoting the growth of several beneficial bacteria and affect the metabolic pathways of intestinal bacteria.Citation97,Citation98 In general, evidence on the effects of NAC treatment on reproductive aging and gut microbiota are rare and fertility-related researches to assess the effects of NAC administration are needed.

Antiaging diet

In addition to antiaging drugs, antiaging diets seem to be a more popular option, of which caloric restriction (CR) is currently regarded as the most reproducible strategy.Citation90,Citation95 CR refers to reducing energy intake without incurring malnutrition or a lack of essential nutrients.Citation93,Citation117 As a dominant paradigm for antiaging diets, its life-extending effect has been widely acknowledged.Citation99 Over the past decades, attention has been given to the effect of CR on reproduction and gut microbiota ().

Table 2. Caloric restriction modulates ovarian function.

Table 3. Gut microbiota alterations induced by caloric restriction.

Obesity, excess body fat and insulin resistance have detrimental effects on ovarian follicle development, thereby damaging the reproductive capacity of female animals.Citation100,Citation101,Citation140–143 CR was associated with weight loss, reduced abdominal visceral fat accumulation, and increased insulin sensitivity.Citation86,Citation144 Consequently, imposed ovarian aging-delay effects through the inhibition of follicle loss and preservation of the follicle pool. Unlike in control rodents, rodents that underwent CR treatment showed significantly increased numbers of primordial follicles and inhibited development of follicles at different stagesCitation86,Citation118,Citation120,Citation122,Citation123,Citation145 as well as an increased ratio of estrogen to androgen receptors.Citation119 Ten weeks of CR feeding decreased serum LH, FSH, and estrogen levels.Citation120 CR exerted a similar effect in HFD-induced POI mice.Citation116 The p53 downregulation induced by CR may inhibit follicle atresia because of its effects on the cell cycle and apoptosis.Citation145 CR can also rescue age-related microbiota alterations observed in aged female mice.Citation121 Three months of 34% CR helped to attain significant weight loss, and decreased waist circumference coincided with changes in five bacterial genera (Subdoligranulum, Collinsella, Parabacteroides, Alistipes, and Bacteroides).Citation146 Thirty days of 40% CR in mice increased the abundances of Lactobacillaceae, Erysipelotrichaceae, Bacteroidaceae and Verrucomicrobiaceae and decreased the abundance of Firmicutes at the family level, improved glucose metabolism, fat browning, and cold tolerance and reduced LPS-binding protein and circulating LPS levels.Citation129 In addition, severe CR decreased body fat deposition and improved glucose toleranceCitation125 but enriched Akkermansia which capable of foraging on host glycans and expensed Roseburia, Ruminococcus, and Eubacterium which specialized for the metabolism of plant polysaccharides in human feces.Citation147

Chronic inflammation is another common condition in agingCitation130 as well as in ovarian aging.Citation38 Inflammation might be one of the mechanisms contributing to reproductive aging. Lliberos et al. reported that the increase in the intraovarian percentage of B cells, CD4+ T cells and macrophages was consistent with the process of ovarian aging and that age-dependent overexpression of multiple proinflammatory cytokines, including IL-6, TNF-α, and IL-1α/β, and the inflammasome genes NLRP3 and ASC was observed during reproductive aging.Citation148 Inhibition of the NLRP3 inflammasome helps to prevent ovarian aging.Citation38 The gut microbiota plays a vital role in the modulation of the inflammatory response.Citation149,Citation150 CR might delay immune senescence by shaping the gut microbiome. CR-microbiota transplantation increased alpha diversity and lowered the abundances of Clostridium ramosum, Hungatella hathewayi, and Alistipes obesi in recipients, and these CR-related microbiota reduced the levels of intestinal and hepatic effector memory CD8+ T cells, intestinal memory B cells, and hepatic effector memory CD4+ T cells.Citation125 Another study discovered five other CR-related operational taxonomic units in obese women: Anaerostipes hadrus, Blautia sp., Agathobacter rectalis, Ruminococcus faecis within Ruminococcaceae, and Bifidobacterium sp. within the family Bifidobacteriaceae, and reduced systemic inflammation was found in obese women after CR.Citation151 Moreover, CR for 2 weeks dramatically reduced intestinal inflammation and increased the protective intestinal microbiota taxon Lactobacillus and the survival rate of 2-month-old female mice after lethal-dose methotrexate exposure.Citation124 The increased Bifidobacterium and Lactobacillus levels induced by lifelong CR were inversely correlated with the levels of bile acids, including ChDxCA and secondary bile acids (DxCA/TDxCA and a ChDxCA derivative).Citation128 The Lactobacillus-dominant microbial community promoted by CR decreased the levels of systemic microbial antigens and inflammatory markers.Citation126 Based on the discovery of several molecular pathways associated with CR mentioned above, CR might be a possible strategy to improve inflammation and possibly promote obesity-related infertility and reproductive span.

Notably, these anti-aging interventions described above either examined the effects on the gut microbiome or ovarian function, and few studies except oneCitation84 speculated on anti-aging drugs, the gut microbiome, and ovarian function simultaneously. Thus, the current evidence that the gut microbiome involved in the mechanism of treatment affects ovarian function is fragmented and scattered, which is also a limitation of this review. There is still a large gap in how these antiaging interventions interact between the gut microbiota and ovarian function. Thus, more research focusing on the effects of these interventions on the gut microbiome and ovarian function is urgently needed to link the interaction of the three parts.

Conclusions

In conclusion, altered composition and function of the gut microbiota play an important role in the pathogenesis of reproductive aging. Experimental and clinical studies have uncovered the relationship between gut dysbiosis and follicle development, as well as a disturbed immune response. Results from FMT studies provide a new insight to anti-ovarian aging, that is the maintenance of youthful gut microbiota helps to preserve ovarian function and prevent ovarian-related diseases. Microbiota-based intervention to delay or reserve ovarian aging is an appealing approach and may offer new therapeutic strategies for intestinal microbiota regulation to improve female fertility.

Furthermore, investigation of antiaging interventions such as antiaging drugs and CR may improve the gut microbial imbalance and promote a healthier intestinal ecological environment. However, evidence from the current scientific literature cannot offer direct conclusions regarding these measures. The majority of the relevant studies were conducted in animal models, which cannot simply apply to human beings. Therefore, future studies should shift from simple correlation analysis to large-scale cohort research and focus on the potential causes and underlying mechanisms to verify the beneficial effects of these interventions in ovarian aging.

Given the wide alterations in the gut microbiota composition and function throughout ovarian aging, it has been suggested that the gut microbiota may be suitable for deciphering the processes of expected and unexpected ovarian aging in women. Imbalance in the gut microbiota may lead to the progression of various ovarian aging-related conditions. Although ovarian aging is unavoidable, maintenance of a balanced gut microbiota is a potential way to delay ovarian aging and subsequent adverse outcomes.

Perspectives

Recent studies have achieved considerable progress in elucidating the roles of the gut microbiota in ovarian aging. However, it is still unclear whether gut microbial changes are the cause or consequence of ovarian aging, and the exact time point at which to modulate the gut microbiota to exert anti-ovarian aging effects remains unsolved. Understanding the relationship between the gut microbiota and ovarian aging is still relatively preliminary, the core mechanism needs to be further explored, and more high-quality evidence is urgently needed. Many factors may affect the gut microbiota, such as age, obesity, dietary pattern, antibiotic usage, and smoking, and these confounders need to be fully considered in future studies. Additionally, metagenomic analyses with a strain-level resolution are warranted because of the high variability of specific species and strains.

Based on the current scientific literature, we can only find some bacterial groups that are closely related to ovarian aging, and the current evidence is relatively scattered. The effects are likely achieved through a pleiotropic mechanism, one of which is an altered production of microbe-associated metabolites.Citation127 The role and molecular mechanism of specific strains and their metabolites in follicle development are still uncertain and remain largely unexplored. Several studies have reported that both intestinal and serum metabolite disorders exist in POI samples,Citation46,Citation152,Citation153 and some metabolites may mediate the function of the hypothalamic-pituitary-ovary axis.Citation152,Citation154 Significant correlations among SCFAs with follicular development and follicular fluid hormones were found.Citation16 Supplementation with specific metabolites may be another choice for regulating ovarian function. Zhang, et al found that polyamine metabolite spermidine level was reduced in ovaries of aged mice and supplementation with spermidine promoted follicle development, rejuvenated oocyte quality and improved fertility rate of aged mice.Citation155 And Guo, et al found that Branch chain amino acid (BCAA) insufficiency could lead to POI, and supplementation with BCAA ameliorated ovarian dysfunction from reactive oxygen species-induced POI in mice.Citation152 Long-term NMN treatment led to significantly higher levels of bile acid-related metabolites.Citation78 However, there are still a series of specific metabolites should be further identified and validated. In addition, follicular atresia is initiated with the apoptosis of GCs after birth.Citation156 A study has indicated that the gut microbiota may participate in regulating ovarian follicular development via SCFAs affecting GC apoptosis.Citation16 The effects of other certain metabolites to GC apoptosis are worthy exploring.

Furthermore, numerous studies have suggested that bacteriotherapy using three slightly different agents: probiotics, prebiotics, and synbiotics are promising for the prevention and treatment of human general aging and age-related disorders,Citation5,Citation103,Citation157–161 in which Lactobacillus and bifidobacterium are the most commonly used, though the effects varied across studies, which depending on dosage, duration, and their components.Citation162 Several clinical trials have demonstrated that the intake of a probiotic mixture decreasing inflammatory levels, increased the abundance of an anti-inflammatory commensal bacterium and improved cognitive function in older people.Citation80,Citation131 Additionally, studies have shown that some probiotic strains have beneficial effects on male reproductive dysfunction.Citation132,Citation133 Supplementation of Lactobacillus and Bifidobacterium improved sperm motility and reduced rate of DNA fragmentation in males.Citation134 Supplementation with probiotics can restore serum testosterone levels and increased spermatogenesis in aging mice.Citation135 However, the effects of the probiotics, prebiotics, and synbiotics on the ovarian aging are unclear, and more studies are required to prove whether they could be effective strategies to counteract ovarian aging.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This work was supported by (1) the National Natural Science Foundation of China (81871141 and 82201781); (2) the National High-Level Hospital Clinical Research Funding (2022-PUMCH-B-123); (3) the National Key Research and Development Program (2018YFC1004801); and (4) the CAMS Innovation Fund for Medical Sciences (CIFMS) (2020-I2M-CT-B-040).

References

  • Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–19. doi:10.1038/s41579-020-0433-9.
  • Hou K, Wu ZX, Chen XY, Wang JQ, Zhang D, Xiao C, Zhu D, Koya JB, Wei L, Li J, et al. Microbiota in health and diseases. Sig Transduct Target Ther. 2022;7(1):135. doi:10.1038/s41392-022-00974-4.
  • Ling Z, Liu X, Cheng Y, Yan X, Wu S. Gut microbiota and aging. Crit Rev Food Sci Nutr. 2022;62(13):3509–3534. doi:10.1080/10408398.2020.1867054.
  • Haran JP, McCormick BA. Aging, frailty, and the microbiome—How dysbiosis Influences human aging and disease. Gastroenterology. 2021;160(2):507–23. doi:10.1053/j.gastro.2020.09.060.
  • Vaiserman AM, Koliada AK, Marotta F. Gut microbiota: a player in aging and a target for anti-aging intervention. Ageing Res Rev. 2017;35:36–45. doi:10.1016/j.arr.2017.01.001.
  • Nie C, Li Y, Li R, Yan Y, Zhang D, Li T, Li Z, Sun Y, Zhen H, Ding J, et al. Distinct biological ages of organs and systems identified from a multi-omics study. Cell Rep. 2022;38(10):110459. doi:10.1016/j.celrep.2022.110459.
  • Wilmanski T, Diener C, Rappaport N, Patwardhan S, Wiedrick J, Lapidus J, Earls JC, Zimmer A, Glusman G, Robinson M, et al. Gut microbiome pattern reflects healthy ageing and predicts survival in humans. Nat Metabolism. 2021;3(2):274–286. doi:10.1038/s42255-021-00348-0.
  • Ke S, Mitchell SJ, MacArthur MR, Kane AE, Sinclair DA, Venable EM, Chadaideh K, Carmody R, Grodstein F, Mitchell J, et al. Gut microbiota predicts healthy late-life aging in male mice. Nutrients. 2021;13(9):3290. doi:10.3390/nu13093290.
  • Parker A, Romano S, Ansorge R, Aboelnour A, Le Gall G, Savva GM, Pontifex MG, Telatin A, Baker D, Jones E, et al. Fecal microbiota transfer between young and aged mice reverses hallmarks of the aging gut, eye, and brain. Microbiome. 2022;10(1):68. doi:10.1186/s40168-022-01243-w.
  • Boehme M, Guzzetta KE, Bastiaanssen TFS, van de Wouw M, Moloney GM, Gual-Grau A, Spichak S, Olavarría-Ramírez L, Fitzgerald P, Morillas E, et al. Microbiota from young mice counteracts selective age-associated behavioral deficits. Nat Aging. 2021;1(8):666–76. doi:10.1038/s43587-021-00093-9.
  • Broekmans FJ, Soules MR, Fauser BC. Ovarian aging: mechanisms and clinical consequences. Endocr Rev. 2009;30(5):465–493. doi:10.1210/er.2009-0006.
  • Li J, Xiong M, Fu XH, Fan Y, Dong C, Sun X, Zheng F, Wang, SW, Liu L, Xu M, et al. Determining a multimodal aging clock in a cohort of Chinese women. Med (New York, NY). 2023;4(11):825–48.
  • Wu M, Lu Z, Zhu Q, Ma L, Xue L, Li Y, Zhou S, Yan W, Ye W, Zhang J, et al. DDX04+ stem cells in the ovaries of postmenopausal women: existence and differentiation potential. Stem Cells (Dayton, Ohio). 2022;40(1):88–101. doi:10.1093/stmcls/sxab002.
  • Wu M, Huang Y, Zhu Q, Zhu X, Xue L, Xiong J, Chen Y, Wu C, Guo Y, Li Y, et al. Adipose tissue and ovarian aging: Potential mechanism and protective strategies. Ageing Res Rev. 2022;80:101683. doi:10.1016/j.arr.2022.101683.
  • Qi X, Yun C, Pang Y, Qiao J. The impact of the gut microbiota on the reproductive and metabolic endocrine system. Gut Microbes. 2021;13(1):1–21. doi:10.1080/19490976.2021.1894070.
  • Xu B, Qin W, Chen Y, Tang Y, Zhou S, Huang J, Ma L, Yan X. Multi-omics analysis reveals gut microbiota-ovary axis contributed to the follicular development difference between Meishan and landrace × Yorkshire sows. J Anim Sci Biotechnol. 2023;14(1):68. doi:10.1186/s40104-023-00865-w.
  • Nelson HD. Menopause. Lancet (London, England). 2008;371(9614):760–70. doi:10.1016/S0140-6736(08)60346-3.
  • Takahashi TA, Johnson KM. Menopause. Med Clin North Am. 2015;99(3):521–534. doi:10.1016/j.mcna.2015.01.006.
  • Al-Azzawi F, Palacios S. Hormonal changes during menopause. Maturitas. 2009;63(2):135–137. doi:10.1016/j.maturitas.2009.03.009.
  • Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas. 2017;103:45–53. doi:10.1016/j.maturitas.2017.06.025.
  • Hu S, Ding Q, Zhang W, Kang M, Ma J, Zhao L. Gut microbial beta-glucuronidase: a vital regulator in female estrogen metabolism. Gut Microbes. 2023;15(1):2236749. doi:10.1080/19490976.2023.2236749.
  • Plottel CS, Blaser MJ. Microbiome and malignancy. Cell Host & Microbe. 2011;10(4):324–335. doi:10.1016/j.chom.2011.10.003.
  • Santos-Marcos JA, Rangel-Zuñiga OA, Jimenez-Lucena R, Quintana-Navarro GM, Garcia-Carpintero S, Malagon MM, Landa BB, Tena-Sempere M, Perez-Martinez P, Lopez-Miranda J, et al. Influence of gender and menopausal status on gut microbiota. Maturitas. 2018;116:43–53. doi:10.1016/j.maturitas.2018.07.008.
  • Yang M, Wen S, Zhang J, Peng J, Shen X, Xu L. Systematic review and meta-analysis: changes of gut microbiota before and after menopause. Dis Markers. 2022;2022:3767373. doi:10.1155/2022/3767373.
  • Kaliannan K, Robertson RC, Murphy K, Stanton C, Kang C, Wang B, Hao L, Bhan AK, Kang JX. Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome. 2018;6(1):205. doi:10.1186/s40168-018-0587-0.
  • Nakatsu CH, Armstrong A, Clavijo AP, Martin BR, Barnes S, Weaver CM, Wong V. Fecal bacterial community changes associated with isoflavone metabolites in postmenopausal women after soy bar consumption. PLoS ONE. 2014;9(10):e108924. doi:10.1371/journal.pone.0108924.
  • Mayneris-Perxachs J, Arnoriaga-Rodríguez M, Luque-Córdoba D, Priego-Capote F, Pérez-Brocal V, Moya A, Burokas A, Maldonado R, Fernández-Real J-M. Gut microbiota steroid sexual dimorphism and its impact on gonadal steroids: influences of obesity and menopausal status. Microbiome. 2020;8(1):136. doi:10.1186/s40168-020-00913-x.
  • Zhao H, Chen J, Li X, Sun Q, Qin P, Wang Q. Compositional and functional features of the female premenopausal and postmenopausal gut microbiota. FEBS Lett. 2019;593(18):2655–2664. doi:10.1002/1873-3468.13527.
  • Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444(7122):1022–1023. doi:10.1038/4441022a.
  • Kasahara K, Krautkramer KA, Org E, Romano KA, Kerby RL, Vivas EI, Mehrabian M, Denu JM, Backhed F, Lusis AJ, et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat microbiol. 2018;3(12):1461–71. doi:10.1038/s41564-018-0272-x.
  • Chen Y, Liu Y, Wang Y, Chen X, Wang C, Chen X, Yuan X, Liu L, Yang J, Zhou X, et al. Prevotellaceae produces butyrate to alleviate PD-1/PD-L1 inhibitor-related cardiotoxicity via PPARα-CYP4X1 axis in colonic macrophages. J Exp Clin Cancer Res. 2022;41(1):1. doi:10.1186/s13046-021-02201-4.
  • Konhilas JP, Sanchez JN, Regan JA, Constantopoulos E, Lopez-Pier M, Cannon DK, Skaria R, McKee LA, Chen H, Lipovka Y, et al. Using 4-vinylcyclohexene diepoxide as a model of menopause for cardiovascular disease. Am J Physiol Heart Circ Physiol. 2020;318(6):H1461–H1473. doi:10.1152/ajpheart.00555.2019.
  • Cao LB, Leung CK, Law PW, Lv Y, Ng CH, Liu HB, Lu G, Ma JL, Chan WY. Systemic changes in a mouse model of VCD-induced premature ovarian failure. Life Sci. 2020;262:118543. doi:10.1016/j.lfs.2020.118543.
  • Biteau B, Hochmuth CE, Jasper H. JNK activity in somatic stem cells causes loss of tissue homeostasis in the aging Drosophila gut. Cell Stem Cell. 2008;3(4):442–455. doi:10.1016/j.stem.2008.07.024.
  • Biteau B, Karpac J, Supoyo S, Degennaro M, Lehmann R, Jasper H, Kim SK. Lifespan extension by preserving proliferative homeostasis in Drosophila. Plos Genet. 2010;6(10):e1001159. doi:10.1371/journal.pgen.1001159.
  • Ahmed SMH, Maldera JA, Krunic D, Paiva-Silva GO, Pénalva C, Teleman AA, Edgar BA. Fitness trade-offs incurred by ovary-to-gut steroid signalling in Drosophila. Nature. 2020;584(7821):415–9. doi:10.1038/s41586-020-2462-y.
  • Xu L, Zhang Q, Dou X, Wang Y, Wang J, Zhou Y, Liu X, Li J. Fecal microbiota transplantation from young donor mice improves ovarian function in aged mice. J Genet Genomics. 2022;49(11):1042–52. doi:10.1016/j.jgg.2022.05.006.
  • Navarro-Pando JM, Alcocer-Gomez E, Castejon-Vega B, Navarro-Villaran E, Condes-Hervas M, Mundi-Roldan M, Muntané J, Pérez-Pulido AJ, Bullon P, Wang C, et al. Inhibition of the NLRP3 inflammasome prevents ovarian aging. Sci Adv. 2021;7(1). doi:10.1126/sciadv.abc7409.
  • Hu C, Liu M, Sun B, Tang L, Zhou X, Chen L. Young fecal transplantation mitigates the toxicity of perfluorobutanesulfonate and potently refreshes the reproductive endocrine system in aged recipients. Environ Int. 2022;167:107418. doi:10.1016/j.envint.2022.107418.
  • Webber L, Davies M, Anderson R, Bartlett J, Braat D, Cartwright B, Cifkova R, de Muinck Keizer-Schrama S, Hogervorst E, Janse F. ESHRE guideline: management of women with premature ovarian insufficiency. Hum Reprod. 2016;31:926–37.
  • Webber L, Anderson RA, Davies M, Janse F, Vermeulen N. HRT for women with premature ovarian insufficiency: a comprehensive review. Human Reprod Open. 2017;2017(2):hox007. doi:10.1093/hropen/hox007.
  • Panay N, Anderson RA, Nappi RE, Vincent AJ, Vujovic S, Webber L, Wolfman W. Premature ovarian insufficiency: an international menopause society white paper. Climacteric. 2020;23(5):426–46. doi:10.1080/13697137.2020.1804547.
  • Georgakis MK, Beskou-Kontou T, Theodoridis I, Skalkidou A, Petridou ET. Surgical menopause in association with cognitive function and risk of dementia: a systematic review and meta-analysis. Psychoneuroendocrinology. 2019;106:9–19. doi:10.1016/j.psyneuen.2019.03.013.
  • Honigberg MC, Zekavat SM, Aragam K, Finneran P, Klarin D, Bhatt DL, Januzzi JL, Scott NS, Natarajan P. Association of premature natural and surgical menopause with incident cardiovascular disease. JAMA. 2019;322(24):2411–21. doi:10.1001/jama.2019.19191.
  • Shuster LT, Rhodes DJ, Gostout BS, Grossardt BR, Rocca WA. Premature menopause or early menopause: long-term health consequences. Maturitas. 2010;65(2):161–166. doi:10.1016/j.maturitas.2009.08.003.
  • Jiang L, Fei H, Tong J, Zhou J, Zhu J, Jin X, Shi Z, Zhou Y, Ma X, Yu H, et al. Hormone replacement therapy reverses gut microbiome and serum metabolome alterations in premature ovarian insufficiency. Front Endocrinol (Lausanne). 2021;12:794496. doi:10.3389/fendo.2021.794496.
  • Wu J, Zhuo Y, Liu Y, Chen Y, Ning Y, Yao J. Association between premature ovarian insufficiency and gut microbiota. BMC Pregnancy Childbirth. 2021;21(1):418. doi:10.1186/s12884-021-03855-w.
  • Ishizuka B. Current understanding of the etiology, symptomatology, and treatment options in Premature Ovarian Insufficiency (POI). Front Endocrinol (Lausanne). 2021;12:626924. doi:10.3389/fendo.2021.626924.
  • Wang Y, Zou Y, Wang W, Zheng Q, Feng Y, Dong H, Tan Z, Zeng X, Zhao Y, Peng D, et al. Knowledge of iatrogenic premature ovarian insufficiency among Chinese obstetricians and gynaecologists: a national questionnaire survey. J Ovarian Res. 2020;13(1):134. doi:10.1186/s13048-020-00739-z.
  • Gargus E, Deans R, Anazodo A, Woodruff TK. Management of primary ovarian insufficiency symptoms in survivors of childhood and adolescent Cancer. J Natl Compr Canc Netw. 2018;16(9):1137–1149. doi:10.6004/jnccn.2018.7023.
  • Cattoni A, Parissone F, Porcari I, Molinari S, Masera N, Franchi M, Cesaro S, Gaudino R, Passoni P, Balduzzi A, et al. Hormonal replacement therapy in adolescents and young women with chemo- or radio-induced premature ovarian insufficiency: practical recommendations. Blood Rev. 2021;45:100730. doi:10.1016/j.blre.2020.100730.
  • van Dorp W, Mulder RL, Kremer LC, Hudson MM, van den Heuvel-Eibrink MM, van den Berg MH, Levine JM, van Dulmen-den Broeder E, di Iorgi N, Albanese A, et al. Recommendations for premature ovarian insufficiency surveillance for female survivors of childhood, adolescent, and young adult cancer: a report from the international late effects of childhood cancer guideline harmonization group in collaboration with the PanCareSurFup consortium. J Clin Oncol. 2016;34(28):3440–50. doi:10.1200/JCO.2015.64.3288.
  • Green DM, Nolan VG, Goodman PJ, Whitton JA, Srivastava D, Leisenring WM, Neglia JP, Sklar CA, Kaste SC, Hudson MM, et al. The cyclophosphamide equivalent dose as an approach for quantifying alkylating agent exposure: a report from the childhood Cancer survivor study. Pediatr Blood Cancer. 2014;61(1):53–67. doi:10.1002/pbc.24679.
  • Chemaitilly W, Li Z, Krasin MJ, Brooke RJ, Wilson CL, Green DM, Klosky JL, Barnes N, Clark KL, Farr JB, et al. Premature ovarian insufficiency in childhood Cancer survivors: a report from the St. Jude Lifetime Cohort. J Clin Endocrinol Metab. 2017;102(7):2242–50. doi:10.1210/jc.2016-3723.
  • van Dorp W, Haupt R, Anderson RA, Mulder RL, van den Heuvel-Eibrink MM, van Dulmen-den BE, Su HI, Winther JF, Hudson MM, Levine JM, et al. Reproductive function and outcomes in female survivors of childhood, adolescent, and young adult Cancer: a review. J Clin Oncol. 2018;36(21):2169–80. doi:10.1200/JCO.2017.76.3441.
  • Tong J, Zhang X, Fan Y, Chen L, Ma X, Yu H, Li J, Guan X, Zhao P, Yang J, et al. Changes of intestinal microbiota in ovarian Cancer patients treated with surgery and chemotherapy. Cancer Manag Res 2020; 12:8125–8135. 10.2147/CMAR.S265205.
  • Medina-Contreras J, Villalobos-Molina R, Zarain-Herzberg A, Balderas-Villalobos J. Ovariectomized rodents as a menopausal metabolic syndrome model. A minireview. Mol Cell Biochem. 2020;475(1–2):261–276. doi:10.1007/s11010-020-03879-4.
  • Vodegel EV, Guler Z, Ras L, Mackova K, Groeneveld A, Bezuidenhout D, Deprest J, Jeffery ST, Roovers JPWR. Vaginal changes after ovariectomy in ewes: a large animal model for genitourinary syndrome of menopause. Int J Gynaecol Obstet. 2023;162(3):1042–9. doi:10.1002/ijgo.14816.
  • Brent MB. Pharmaceutical treatment of bone loss: from animal models and drug development to future treatment strategies. Pharmacology & Therapeutics. 2023;244:108383. doi:10.1016/j.pharmthera.2023.108383.
  • Diaz Brinton R. Minireview: translational animal models of human menopause: challenges and emerging opportunities. Endocrinology. 2012;153(8):3571–3578. doi:10.1210/en.2012-1340.
  • Xiao HH, Lu L, Poon CC, Chan CO, Wang LJ, Zhu YX, Zhou L-P, Cao S, Yu W-X, Wong KY, et al. The lignan-rich fraction from Sambucus Williamsii Hance ameliorates dyslipidemia and insulin resistance and modulates gut microbiota composition in ovariectomized rats. Biomed Pharmacother. 2021;137:111372. doi:10.1016/j.biopha.2021.111372.
  • Tu MY, Han KY, Chang GR, Lai GD, Chang KY, Chen CF, Lai J-C, Lai C-Y, Chen H-L, Chen C-M, et al. Kefir peptides prevent estrogen deficiency-induced bone loss and modulate the structure of the gut microbiota in ovariectomized mice. Nutrients. 2020;12(11):12. doi:10.3390/nu12113432.
  • Zhang Z, Chen Y, Xiang L, Wang Z, Xiao GG, Hu J. Effect of curcumin on the diversity of gut microbiota in ovariectomized rats. Nutrients. 2017;9(10):1146. doi:10.3390/nu9101146.
  • Huang J, Shan W, Li F, Wang Z, Cheng J, Lu F, Guo E, Beejadhursing R, Xiao R, Liu C, et al. Fecal microbiota transplantation mitigates vaginal atrophy in ovariectomized mice. Aging (Albany NY). 2021;13(5):7589–7607. doi:10.18632/aging.202627.
  • Zhang YW, Cao MM, Li YJ, Lu PP, Dai GC, Zhang M, Wang H, Rui Y-F. Fecal microbiota transplantation ameliorates bone loss in mice with ovariectomy-induced osteoporosis via modulating gut microbiota and metabolic function. J Orthop Translat. 2022;37:46–60. doi:10.1016/j.jot.2022.08.003.
  • Chevalier C, Kieser S, Çolakoğlu M, Hadadi N, Brun J, Rigo D, Suárez-Zamorano N, Spiljar M, Fabbiano S, Busse B, et al. Warmth prevents bone loss through the gut microbiota. Cell Metab. 2020;32(4):575–90.e7. doi:10.1016/j.cmet.2020.08.012.
  • Yuan Y, Yang J, Zhuge A, Li L, Ni S. Gut microbiota modulates osteoclast glutathione synthesis and mitochondrial biogenesis in mice subjected to ovariectomy. Cell Prolif. 2022;55(3):e13194. doi:10.1111/cpr.13194.
  • Lee HJ, Park MJ, Joo BS, Joo JK, Kim YH, Yang SW, Kim C-W, Kim KH. Effects of coenzyme Q10 on ovarian surface epithelium-derived ovarian stem cells and ovarian function in a 4-vinylcyclohexene diepoxide-induced murine model of ovarian failure. Reprod Biol Endocrinol. 2021;19(1):59. doi:10.1186/s12958-021-00736-x.
  • Secomandi L, Borghesan M, Velarde M, Demaria M. The role of cellular senescence in female reproductive aging and the potential for senotherapeutic interventions. Hum Reprod Update. 2022;28(2):172–189. doi:10.1093/humupd/dmab038.
  • Wang L, Chen Y, Wei J, Guo F, Li L, Han Z, Wang Z, Zhu H, Zhang X, Li Z, et al. Administration of nicotinamide mononucleotide improves oocyte quality of obese mice. Cell Prolif. 2022;55(11):e13303. doi:10.1111/cpr.13303.
  • Hao EY, Chen H, Wang DH, Huang CX, Tong YG, Chen YF, Zhou R-Y, Huang R-L. Melatonin regulates the ovarian function and enhances follicle growth in aging laying hens via activating the mammalian target of rapamycin pathway. Poult Sci. 2020;99(4):2185–95. doi:10.1016/j.psj.2019.11.040.
  • Wang Y, Fan L, Huang J, Liang J, Wang X, Ren Y, Li H, Yue T, Gao Z. Evaluation of chemical composition, antioxidant activity, and gut microbiota associated with pumpkin juice fermented by rhodobacter sphaeroides. Food Chem. 2023;401:134122. doi:10.1016/j.foodchem.2022.134122.
  • Zhang L, Zhang Z, Wang J, Lv D, Zhu T, Wang F, Tian X, Yao Y, Ji P, Liu G, 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(3):e12550. doi:10.1111/jpi.12550.
  • Yan F, Zhao Q, Li Y, Zheng Z, Kong X, Shu C, Liu Y, Shi Y. The role of oxidative stress in ovarian aging: a review. J Ovarian Res. 2022;15(1):100. doi:10.1186/s13048-022-01032-x.
  • Wang J, Jia R, Gong H, Celi P, Zhuo Y, Ding X, Bai S, Zeng Q, Yin H, Xu S, et al. The effect of oxidative stress on the chicken ovary: involvement of microbiota and Melatonin interventions. Antioxidants (Basel, Switzerland). 2021;10(9):10. doi:10.3390/antiox10091422.
  • Tamura H, Jozaki M, Tanabe M, Shirafuta Y, Mihara Y, Shinagawa M, Tamura I, Maekawa R, Sato S, Taketani T, et al. Importance of Melatonin in assisted reproductive technology and ovarian aging. Int J Mol Sci. 2020;21(3):21. doi:10.3390/ijms21031135.
  • Ivanova AY, Shirokov IV, Toshchakov SV, Kozlova AD, Obolenskaya ON, Mariasina SS, Ivlev VA, Gartseev IB, Medvedev OS. Effects of coenzyme Q10 on the biomarkers (hydrogen, methane, SCFA and TMA) and composition of the gut microbiome in rats. Pharmaceuticals (Basel, Switzerland). 2023;16(5):686. doi:10.3390/ph16050686.
  • Huang P, Jiang A, Wang X, Zhou Y, Tang W, Ren C, Qian X, Zhou Z, Gong A. NMN maintains intestinal homeostasis by regulating the gut microbiota. Front Nutr. 2021;8:714604. doi:10.3389/fnut.2021.714604.
  • Zhu H, Li X, Qiao M, Sun X, Li G, Anderson RM. Resveratrol alleviates inflammation and ER stress through SIRT1/NRF2 to delay ovarian aging in a short-lived fish. J Gerontol A Biol Sci Med Sci. 2023;78(4):596–602. doi:10.1093/gerona/glad009.
  • Spaiser SJ, Culpepper T, Nieves C Jr., Ukhanova M, Mai V, Percival SS, Christman MC, Langkamp-Henken B. Lactobacillus gasseri KS-13, Bifidobacterium bifidum G9-1, and Bifidobacterium longum MM-2 ingestion induces a less inflammatory cytokine profile and a potentially beneficial shift in gut microbiota in older adults: a randomized, Double-blind, placebo-controlled, crossover study. J Am Coll Nutr. 2015;34(6):459–69. doi:10.1080/07315724.2014.983249.
  • Bertoldo MJ, Listijono DR, Ho WJ, Riepsamen AH, Goss DM, Richani D, Jin XL, Mahbub S, Campbell JM, Habibalahi A, et al. NAD(+) repletion rescues female fertility during reproductive aging. Cell Rep. 2020;30(6):1670–81.e7. doi:10.1016/j.celrep.2020.01.058.
  • Mourad MM, Shahin SA, El-Ratel IT, El Basuini MF. Effect of treating eggs with coenzyme Q10 (CoQ10) on growth variables, histomorphometry, and antioxidant capacity in Red tilapia (Oreochromis aureus × Oreochromis mossambicus) larvae. Animals: An Open Access J MDPI. 2022;12(17):12. doi:10.3390/ani12172219.
  • He N, Shen G, Jin X, Li H, Wang J, Xu L, Chen J, Cao X, Fu C, Shi D, et al. Resveratrol suppresses microglial activation and promotes functional recovery of traumatic spinal cord via improving intestinal microbiota. Pharmacol Res. 2022;183:106377. doi:10.1016/j.phrs.2022.106377.
  • Wang J, Jia R, Celi P, Zhuo Y, Ding X, Zeng Q, Bai S, Xu S, Yin H, Lv L, et al. Resveratrol alleviating the ovarian function under oxidative stress by alternating microbiota related tryptophan-kynurenine pathway. Front Immunol. 2022;13:911381. doi:10.3389/fimmu.2022.911381.
  • Zhang Y, Zhang J, Wang S. The role of rapamycin in healthspan extension via the delay of Organ aging. Ageing Res Rev. 2021;70:101376. doi:10.1016/j.arr.2021.101376.
  • Garcia DN, Saccon TD, Pradiee J, Rincon JAA, Andrade KRS, Rovani MT, Mondadori RG, Cruz LAX, Barros CC, Masternak MM, et al. Effect of caloric restriction and rapamycin on ovarian aging in mice. Geroscience. 2019;41(4):395–408. doi:10.1007/s11357-019-00087-x.
  • Dou X, Sun Y, Li J, Zhang J, Hao D, Liu W, Wu R, Kong F, Peng X, Li J, et al. Short-term rapamycin treatment increases ovarian lifespan in young and middle-aged female mice. Aging Cell. 2017;16(4):825–836. doi:10.1111/acel.12617.
  • Zhou L, Xie Y, Li S, Liang Y, Qiu Q, Lin H, Zhang Q. Rapamycin prevents cyclophosphamide-induced over-activation of primordial follicle pool through PI3K/Akt/mTOR Signaling pathway in vivo. J Ovarian Res. 2017;10(1):56. doi:10.1186/s13048-017-0350-3.
  • Bitto A, Ito TK, Pineda VV, LeTexier NJ, Huang HZ, Sutlief E, Tung H, Vizzini N, Chen B, Smith K, et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. eLife. 2016;5:e16351. doi:10.7554/eLife.16351.
  • Duan H, Pan J, Guo M, Li J, Yu L, Fan L. Dietary strategies with anti-aging potential: dietary patterns and supplements. Food Res Int. 2022;158:111501. doi:10.1016/j.foodres.2022.111501.
  • Cao S, Yan H, Tang W, Zhang H, Liu J. Effects of dietary coenzyme Q10 supplementation during gestation on the embryonic survival and reproductive performance of high-parity sows. J Anim Sci Biotechnol. 2023;14(1):75. doi:10.1186/s40104-023-00879-4.
  • Schinaman JM, Rana A, Ja WW, Clark RI, Walker DW. Rapamycin modulates tissue aging and lifespan independently of the gut microbiota in Drosophila. Sci Rep. 2019;9(1):7824. doi:10.1038/s41598-019-44106-5.
  • Zheng X, Wang S, Jia W. Calorie restriction and its impact on gut microbial composition and global metabolism. Front Med. 2018;12(6):634–644. doi:10.1007/s11684-018-0670-8.
  • Raghu G, Berk M, Campochiaro PA, Jaeschke H, Marenzi G, Richeldi L, Wen F-Q, Nicoletti F, Calverley PMA. The multifaceted therapeutic role of N-Acetylcysteine (NAC) in disorders characterized by oxidative stress. Curr Neuropharmacol. 2021;19(8):1202–24. doi:10.2174/1570159X19666201230144109.
  • Lee MB, Hill CM, Bitto A, Kaeberlein M. Antiaging diets: separating fact from fiction. Sci. 2021;374(6570):eabe7365. doi:10.1126/science.abe7365.
  • Liu J, Liu M, Ye X, Liu K, Huang J, Wang L, Ji G, Liu N, Tang X, Baltz JM, et al. Delay in oocyte aging in mice by the antioxidant N-acetyl-L-cysteine (NAC). Hum Reprod. 2012;27(5):1411–20. doi:10.1093/humrep/des019.
  • Zheng J, Yuan X, Zhang C, Jia P, Jiao S, Zhao X, Yin H, Du Y, Liu H. N-Acetylcysteine alleviates gut dysbiosis and glucose metabolic disorder in high-fat diet-fed mice. J Diabetes. 2019;11(1):32–45. doi:10.1111/1753-0407.12795.
  • Ding Q, Guo R, Pei L, Lai S, Li J, Yin Y, Xu T, Yang W, Song Q, Han Q, et al. N-Acetylcysteine alleviates high fat diet-induced hepatic steatosis and liver injury via regulating the intestinal microecology in mice. Food Funct. 2022;13(6):3368–3380. doi:10.1039/D1FO03952K.
  • Giacomello E, Toniolo L. The potential of calorie restriction and calorie restriction mimetics in delaying aging: focus on experimental models. Nutrients. 2021;13(7):2346. doi:10.3390/nu13072346.
  • Liu T, Lin J, Chen C, Nie X, Dou F, Chen J, Wang Z, Gong Z. MicroRNA-146b-5p overexpression attenuates premature ovarian failure in mice by inhibiting the Dab2ip/Ask1/p38-mapk pathway and γH2A.X phosphorylation. Cell Prolif. 2021;54(1):e12954. doi:10.1111/cpr.12954.
  • Liu T, Jing F, Huang P, Geng Z, Xu J, Li J, Chen D, Zhu Y, Wang Z, Huang W, et al. Thymopentin alleviates premature ovarian failure in mice by activating YY2/Lin28A and inhibiting the expression of let-7 family microRnas. Cell Proliferation. 2021;54(8):e13089. doi:10.1111/cpr.13089.
  • Du Y, Gao Y, Zeng B, Fan X, Yang D, Yang M. Effects of anti-aging interventions on intestinal microbiota. Gut Microbes. 2021;13(1):1994835. doi:10.1080/19490976.2021.1994835.
  • Ben-Meir A, Burstein E, Borrego-Alvarez A, Chong J, Wong E, Yavorska T, Naranian T, Chi M, Wang Y, Bentov Y, et al. Coenzyme Q10 restores oocyte mitochondrial function and fertility during reproductive aging. Aging Cell. 2015;14(5):887–895. doi:10.1111/acel.12368.
  • Xu Y, Nisenblat V, Lu C, Li R, Qiao J, Zhen X, Wang S. Pretreatment with coenzyme Q10 improves ovarian response and embryo quality in low-prognosis young women with decreased ovarian reserve: a randomized controlled trial. Reprod Biol Endocrinol. 2018;16(1):29. doi:10.1186/s12958-018-0343-0.
  • Sharideh H, Zhandi M, Zeinoaldini S, Zaghari M, Sadeghi M, Akhlaghi A, Peebles ED. Beneficial effects of dietary coenzyme Q10 on the productive and reproductive variables of broiler breeder hens. Anim Reprod Sci. 2020;213:106256. doi:10.1016/j.anireprosci.2019.106256.
  • Florou P, Anagnostis P, Theocharis P, Chourdakis M, Goulis DG. Does coenzyme Q(10) supplementation improve fertility outcomes in women undergoing assisted reproductive technology procedures? A systematic review and meta-analysis of randomized-controlled trials. J Assist Reprod Genet. 2020;37(10):2377–87. doi:10.1007/s10815-020-01906-3.
  • Nadeeshani H, Li J, Ying T, Zhang B, Lu J. Nicotinamide mononucleotide (NMN) as an anti-aging health product - promises and safety concerns. J Adv Res. 2022;37:267–78. doi:10.1016/j.jare.2021.08.003.
  • Liu S, Kang W, Mao X, Ge L, Du H, Li J, Hou L, Liu D, Yin Y, Liu Y, et al. Melatonin mitigates aflatoxin B1-induced liver injury via modulation of gut microbiota/intestinal FXR/liver TLR4 signaling axis in mice. J Pineal Res. 2022;73(2):e12812. doi:10.1111/jpi.12812.
  • Yin J, Li Y, Han H, Chen S, Gao J, Liu G, Wu X, Deng J, Yu Q, Huang X, et al. Melatonin reprogramming of gut microbiota improves lipid dysmetabolism in high-fat diet-fed mice. J Pineal Res. 2018;65(4):e12524. doi:10.1111/jpi.12524.
  • Huang P, Zhou Y, Tang W, Ren C, Jiang A, Wang X, Qian X, Zhou Z, Gong A. Long-term treatment of nicotinamide mononucleotide improved age-related diminished ovary reserve through enhancing the mitophagy level of granulosa cells in mice. J Nutr Biochem. 2022;101:108911. doi:10.1016/j.jnutbio.2021.108911.
  • Liu M, Yin Y, Ye X, Zeng M, Zhao Q, Keefe DL, Liu L. Resveratrol protects against age-associated infertility in mice. Hum Reprod. 2013;28(3):707–17. doi:10.1093/humrep/des437.
  • Özcan P, Fıçıcıoğlu C, Yıldırım ÖK, Özkan F, Akkaya H, Aslan İ. Protective effect of resveratrol against oxidative damage to ovarian reserve in female Sprague–Dawley rats. Reprod Biomed Online. 2015;31(3):404–10. doi:10.1016/j.rbmo.2015.06.007.
  • Wang P, Li D, Ke W, Liang D, Hu X, Chen F. Resveratrol-induced gut microbiota reduces obesity in high-fat diet-fed mice. Int J Obes. 2020;44(1):213–25. doi:10.1038/s41366-019-0332-1.
  • Guo J, Zhang T, Guo Y, Sun T, Li H, Zhang X, Yin H, Cao G, Yin Y, Wang H, et al. Oocyte stage-specific effects of MTOR determine granulosa cell fate and oocyte quality in mice. Proc Natl Acad Sci USA. 2018;115(23):E5326–e33. doi:10.1073/pnas.1800352115.
  • Rehnitz J, Messmer B, Bender U, Nguyen XP, Germeyer A, Hinderhofer K, Strowitzki T, Capp E. Activation of AKT/mammalian target of rapamycin signaling in the peripheral blood of women with premature ovarian insufficiency and its correlation with FMR1 expression. Reprod Biol Endocrinol. 2022;20(1):44. doi:10.1186/s12958-022-00919-0.
  • Zhou XL, Xu JJ, Ni YH, Chen XC, Zhang HX, Zhang XM, Liu W-J, Luo L-L, Fu Y-C. SIRT1 activator (SRT1720) improves the follicle reserve and prolongs the ovarian lifespan of diet-induced obesity in female mice via activating SIRT1 and suppressing mTOR signaling. J Ovarian Res. 2014;7(1):97. doi:10.1186/s13048-014-0097-z.
  • Duan H, Li J, Yu L, Fan L. The road ahead of dietary restriction on anti-aging: focusing on personalized nutrition. Crit Rev Food Sci Nutr. 2022:1–18. doi:10.1080/10408398.2022.2110034.
  • Isola JVV, Zanini BM, Hense JD, Alvarado-Rincon JA, Garcia DN, Pereira GC, Vieira AD, Oliveira TL, Collares T, Gasperin BG, 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.
  • Słuczanowska-Głąbowska S, Laszczyńska M, Piotrowska K, Grabowska M, Grymuła K, Ratajczak MZ. Caloric restriction increases ratio of estrogen to androgen receptors expression in murine ovaries–potential therapeutic implications. J Ovarian Res. 2015;8(1):57. doi:10.1186/s13048-015-0185-8.
  • Li L, Fu YC, Xu JJ, Lin XH, Chen XC, Zhang XM, Luo L-L. Caloric restriction promotes the reserve of follicle pool in adult female rats by inhibiting the activation of mammalian target of rapamycin signaling. Reprod Sci. 2015;22(1):60–7. doi:10.1177/1933719114542016.
  • Cox LM, Schafer MJ, Sohn J, Vincentini J, Weiner HL, Ginsberg SD, Blaser MJ. Calorie restriction slows age-related microbiota changes in an Alzheimer’s disease model in female mice. Sci Rep. 2019;9(1):17904. doi:10.1038/s41598-019-54187-x.
  • Li L, Fu YC, Xu JJ, Chen XC, Lin XH, Luo LL. Caloric restriction promotes the reproductive capacity of female rats via modulating the level of insulin-like growth factor-1 (IGF-1). Gen Comp Endocr. 2011;174(2):232–237. doi:10.1016/j.ygcen.2011.09.005.
  • Luo LL, Chen XC, Fu YC, Xu JJ, Li L, Lin XH, Xiang Y-F, Zhang X-M. The effects of caloric restriction and a high-fat diet on ovarian lifespan and the expression of SIRT1 and SIRT6 proteins in rats. Aging Clin Exp Res. 2012;24(2):125–33. doi:10.1007/BF03654792.
  • Tang D, Zeng T, Wang Y, Cui H, Wu J, Zou B, Tao Z, Zhang L, Garside GB, Tao S, et al. Dietary restriction increases protective gut bacteria to rescue lethal methotrexate-induced intestinal toxicity. Gut Microbes. 2020;12(1):1714401. doi:10.1080/19490976.2020.1714401.
  • Sbierski-Kind J, Grenkowitz S, Schlickeiser S, Sandforth A, Friedrich M, Kunkel D, Glauben R, Brachs S, Mai K, Thürmer A, et al. Effects of caloric restriction on the gut microbiome are linked with immune senescence. Microbiome. 2022;10(1):57. doi:10.1186/s40168-022-01249-4.
  • Pan F, Zhang L, Li M, Hu Y, Zeng B, Yuan H, Zhao L, Zhang C. Predominant gut Lactobacillus murinus strain mediates anti-inflammaging effects in calorie-restricted mice. Microbiome. 2018;6(1):54. doi:10.1186/s40168-018-0440-5.
  • Krautkramer KA, Fan J, Bäckhed F. Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol. 2021;19(2):77–94. doi:10.1038/s41579-020-0438-4.
  • Kok DEG, Rusli F, van der Lugt B, Lute C, Laghi L, Salvioli S, Picone G, Franceschi C, Smidt H, Vervoort J, et al. Lifelong calorie restriction affects indicators of colonic health in aging C57Bl/6J mice. J Nutr Biochem. 2018;56:152–164. doi:10.1016/j.jnutbio.2018.01.001.
  • Fabbiano S, Suarez-Zamorano N, Chevalier C, Lazarevic V, Kieser S, Rigo D, Leo S, Veyrat-Durebex C, Gaïa N, Maresca M, et al. Functional gut microbiota remodeling contributes to the caloric restriction-induced metabolic Improvements. Cell Metab. 2018;28(6):907–921.e7. doi:10.1016/j.cmet.2018.08.005.
  • López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186(2):243–278. doi:10.1016/j.cell.2022.11.001.
  • Finamore A, Roselli M, Donini L, Brasili DE, Rami R, Carnevali P, Mistura L, Pinto A, Giusti A, Mengheri E, et al. Supplementation with Bifidobacterium longum Bar33 and Lactobacillus helveticus Bar13 mixture improves immunity in elderly humans (over 75 years) and aged mice. Nutrition (Burbank, Los Angeles County, Calif). 2019;63-64:184–192. doi:10.1016/j.nut.2019.02.005.
  • Dardmeh F, Alipour H, Gazerani P, van der Horst G, Brandsborg E, Nielsen HI, Fam B. Lactobacillus rhamnosus PB01 (DSM 14870) supplementation affects markers of sperm kinematic parameters in a diet-induced obesity mice model. Plos One. 2017;12(10):e0185964. doi:10.1371/journal.pone.0185964.
  • Barbonetti A, Cinque B, Vassallo MR, Mineo S, Francavilla S, Cifone MG, Francavilla F. Effect of vaginal probiotic lactobacilli on in vitro–induced sperm lipid peroxidation and its impact on sperm motility and viability. Fertil Steril. 2011;95(8):2485–8. doi:10.1016/j.fertnstert.2011.03.066.
  • Valcarce DG, Genovés S, Riesco MF, Martorell P, Herráez MP, Ramón D, Robles V. Probiotic administration improves sperm quality in asthenozoospermic human donors. Benef Microbes. 2017;8(2):193–206. doi:10.3920/BM2016.0122.
  • Poutahidis T, Springer A, Levkovich T, Qi P, Varian BJ, Lakritz JR, Ibrahim YM, Chatzigiagkos A, Alm EJ, Erdman SE, et al. Probiotic microbes sustain youthful serum testosterone levels and testicular size in aging mice. Plos One. 2014;9(1):e84877. doi:10.1371/journal.pone.0084877.
  • Song C, Peng W, Yin S, Zhao J, Fu B, Zhang J, Mao T, Wu H, Zhang Y. Melatonin improves age-induced fertility decline and attenuates ovarian mitochondrial oxidative stress in mice. Sci Rep. 2016;6(1):35165. doi:10.1038/srep35165.
  • Wang P, Gao J, Ke W, Wang J, Li D, Liu R, Jia Y, Wang X, Chen X, Chen F, et al. Resveratrol reduces obesity in high-fat diet-fed mice via modulating the composition and metabolic function of the gut microbiota. Free Radic Biol Med. 2020;156:83–98. doi:10.1016/j.freeradbiomed.2020.04.013.
  • Anderson EM, Rozowsky JM, Fazzone BJ, Schmidt EA, Stevens BR, O’Malley KA, Scali ST, Berceli SA. Temporal dynamics of the intestinal microbiome following short-term dietary restriction. Nutrients. 2022;14(14):14. doi:10.3390/nu14142785.
  • Fraumene C, Manghina V, Cadoni E, Marongiu F, Abbondio M, Serra M, Palomba A, Tanca A, Laconi E, Uzzau S, et al. Caloric restriction promotes rapid expansion and long-lasting increase of Lactobacillus in the rat fecal microbiota. Gut Microbes. 2018;9(2):104–114. doi:10.1080/19490976.2017.1371894.
  • Long X, Yang Q, Qian J, Yao H, Yan R, Cheng X, Zhang Q, Gu C, Gao F, Wang H, et al. Obesity modulates cell-cell interactions during ovarian folliculogenesis. iScience. 2022;25(1):103627. doi:10.1016/j.isci.2021.103627.
  • Fan Z, Zhang X, Shang Y, Zou M, Zhou M, Q E, Fei S, Chen W, Li J, Zhang X, et al. Intestinal flora changes induced by a high-fat diet promote activation of primordial follicles through macrophage infiltration and inflammatory factor secretion in Mouse ovaries. Int J Mol Sci. 2022;23(9):23. doi:10.3390/ijms23094797.
  • Di Berardino C, Peserico A, Capacchietti G, Zappacosta A, Bernabo N, Russo V, Mauro A, El Khatib M, Gonnella F, Konstantinidou F, et al. High-fat diet and female fertility across lifespan: a comparative lesson from mammal models. Nutrients. 2022;14(20):14. doi:10.3390/nu14204341.
  • Wang N, Luo LL, Xu JJ, Xu MY, Zhang XM, Zhou XL, Liu W-J, Fu Y-C. Obesity accelerates ovarian follicle development and follicle loss in rats. Metabolism. 2014;63(1):94–103. doi:10.1016/j.metabol.2013.09.001.
  • Kendel Jovanovic G, Mrakovcic-Sutic I, Pavicic Zezelj S, Susa B, Rahelic D, Klobucar Majanovic S. The efficacy of an energy-restricted anti-inflammatory diet for the management of obesity in younger adults. Nutrients. 2020;12(11):12. doi:10.3390/nu12113583.
  • Liu WJ, Zhang XM, Wang N, Zhou XL, Fu YC, Luo LL. Calorie restriction inhibits ovarian follicle development and follicle loss through activating SIRT1 signaling in mice. Eur J Med Res. 2015;20(1):22. doi:10.1186/s40001-015-0114-8.
  • Stanislawski MA, Frank DN, Borengasser SJ, Ostendorf DM, Ir D, Jambal P, Bing K, Wayland L, Siebert JC, Bessesen DH, et al. The gut microbiota during a behavioral weight loss intervention. Nutrients. 2021;13(9):3248. doi:10.3390/nu13093248.
  • von Schwartzenberg RJ, Bisanz JE, Lyalina S, Spanogiannopoulos P, Ang QY, Cai J, Dickmann S, Friedrich M, Liu S-Y, Collins SL, et al. Caloric restriction disrupts the microbiota and colonization resistance. Nature. 2021;595(7866):272–7. doi:10.1038/s41586-021-03663-4.
  • Lliberos C, Liew SH, Zareie P, La Gruta NL, Mansell A, Hutt K. Evaluation of inflammation and follicle depletion during ovarian ageing in mice. Sci Rep. 2021;11(1):278. doi:10.1038/s41598-020-79488-4.
  • Cai J, Sun L, Gonzalez FJ. Gut microbiota-derived bile acids in intestinal immunity, inflammation, and tumorigenesis. Cell Host & Microbe. 2022;30(3):289–300. doi:10.1016/j.chom.2022.02.004.
  • Mou Y, Du Y, Zhou L, Yue J, Hu X, Liu Y, Chen S, Lin X, Zhang G, Xiao H, et al. Gut microbiota interact with the brain through systemic chronic inflammation: implications on neuroinflammation, neurodegeneration, and aging. Front Immunol. 2022;13:796288. doi:10.3389/fimmu.2022.796288.
  • Ott B, Skurk T, Hastreiter L, Lagkouvardos I, Fischer S, Buttner J, Kellerer T, Clavel T, Rychlik M, Haller D, et al. Effect of caloric restriction on gut permeability, inflammation markers, and fecal microbiota in obese women. Sci Rep. 2017;7(1):11955. doi:10.1038/s41598-017-12109-9.
  • Guo X, Zhu Y, Guo L, Qi Y, Liu X, Wang J, Zhang J, Cui L, Shi Y, Wang Q, et al. BCAA insufficiency leads to premature ovarian insufficiency via ceramide-induced elevation of ROS. EMBO Mol Med. 2023;15(4):e17450. doi:10.15252/emmm.202317450.
  • Zheng H, Liang X, Zhou H, Zhou T, Liu X, Duan J, Duan J-A, Zhu Y. Integrated gut microbiota and fecal metabolome analyses of the effect of Lycium barbarum polysaccharide on D-galactose-induced premature ovarian insufficiency. Food Funct. 2023;14(15):7209–21. doi:10.1039/D3FO01659E.
  • Qi X, Yun C, Sun L, Xia J, Wu Q, Wang Y, Wang L, Zhang Y, Liang X, Wang L, et al. Gut microbiota–bile acid–interleukin-22 axis orchestrates polycystic ovary syndrome. Nat Med. 2019;25(8):1225–33. doi:10.1038/s41591-019-0509-0.
  • Zhang Y, Bai J, Cui Z, Li Y, Gao Q, Miao Y, Xiong B. Polyamine metabolite spermidine rejuvenates oocyte quality by enhancing mitophagy during female reproductive aging. Nat Aging. 2023;3(11):1372–86. doi:10.1038/s43587-023-00498-8.
  • Shan X, Yu T, Yan X, Wu J, Fan Y, Guan X, Fang F, Lin Y, Zhang Y, Li Y, et al. Proteomic analysis of healthy and atretic porcine follicular granulosa cells. J Proteomics. 2021;232:104027. doi:10.1016/j.jprot.2020.104027.
  • Sharma R, Padwad Y. Probiotic bacteria as modulators of cellular senescence: emerging concepts and opportunities. Gut Microbes. 2020;11(3):335–349. doi:10.1080/19490976.2019.1697148.
  • Xu HY, Li QC, Zhou WJ, Zhang HB, Chen ZX, Peng N, Gong S-Y, Liu B, Zeng F. Anti-oxidative and anti-aging effects of probiotic Fermented Ginseng by modulating gut microbiota and metabolites in Caenorhabditis elegans. Plant Foods Human Nutr (Dordrecht, Netherlands). 2023;78(2):320–8. doi:10.1007/s11130-023-01055-9.
  • Ishaq M, Khan A, Bacha AS, Shah T, Hanif A, Ahmad AA, Ke W, Li F, Ud Din A, Ding Z, et al. Microbiota targeted interventions of probiotic Lactobacillus as an anti-ageing approach: a review. Antioxidants (Basel, Switzerland). 2021;10(12):10. doi:10.3390/antiox10121930.
  • Lew LC, Hor YY, Jaafar MH, Lau ASY, Ong JS, Chuah LO, Yap KP, Azzam G, Azlan A, Liong MT, et al. Lactobacilli modulated AMPK activity and prevented telomere shortening in ageing rats. Benef Microbes. 2019;10(8):883–892. doi:10.3920/BM2019.0058.
  • Mounir M, Ibijbijen A, Farih K, Rabetafika HN, Razafindralambo HL. Synbiotics and their antioxidant properties, mechanisms, and benefits on human and animal health: a narrative review. Biomolecules. 2022;12(10):12. doi:10.3390/biom12101443.
  • Lim MY, Nam YD. Gut microbiome in healthy aging versus those associated with frailty. Gut Microbes. 2023;15(2):2278225. doi:10.1080/19490976.2023.2278225.