- Review
- Open access
- Published:
The risk factors, pathogenesis and treatment of premature ovarian insufficiency
Journal of Ovarian Research volume 18, Article number: 134 (2025)
Abstract
Premature ovarian insufficiency (POI) is a prevalent condition that impacts female reproductive health and overall well-being. It is characterized by ovarian dysfunction, estrogen deficiency, and increased gonadotropin levels in women under 40 years old. The exact etiology and pathogenesis of POI remain incompletely understood, posing significant challenges for its treatment and prevention. This review article examines the pathogenic factors and mechanisms involved in POI, with a particular focus on the impact of environmental toxicants such as atmospheric particulate matter, endocrine-disrupting chemicals, pesticides, microplastics, heavy metals, and cigarette smoke on the development of POI. Furthermore, the treatments for POI are outlined, encompassing hormone replacement therapy, stem cell and exosome therapy, melatonin therapy, traditional Chinese medicine, in vitro activation, platelet-rich plasma therapy, and ovarian tissue cryopreservation. The primary objective is to raise awareness about the potential detrimental effects of environmental toxicant exposure on ovarian function and reserve, urging individuals to minimize their exposure to harmful substances and advocate for environmental protection. This review also aims to serve as a valuable resource for the prevention and management of POI.
Introduction
Premature ovarian insufficiency (POI) is a gynecological endocrine disorder characterized by menstrual irregularities, infertility, elevated serum gonadotropins, and low estradiol levels in women younger than 40 years old [1]. The global incidence of POI is estimated to be around 3.7% [2], and it significantly affects female reproductive capacity and leads to severe health implications [3]. Studies have shown that women with POI face an increased risk of osteoporosis, cardiovascular disease, mental health disorders, and type 2 diabetes [4,5,6,7]. Additionally, POI not only diminishes oocyte quality and quantity but also correlates with reduced clinical pregnancy rates and heightened miscarriage risks [8]. Due to its complexities, the exact etiology and pathogenesis of POI, particularly the involvement of environmental toxicants (ETs) in its onset and progression, remain incompletely understood. This article aims to comprehensively examine the etiology and pathological mechanisms of POI, with a specific focus on the interplay between ETs exposure and POI. Furthermore, it consolidates various treatment modalities for POI to offer insights for its management and prevention.
Etiology of POI
The etiology of POI is heterogeneous, with a significant portion of cases remaining unidentified. Recognized and prevalent causes of POI encompass genetic, iatrogenic, autoimmune, infectious diseases, and environment [9]. Besides idiopathic influences, genetic factors are deemed primary contributors to POI, constituting approximately 20–25% [10]. Turner syndrome and fragile X premutation, stemming from X chromosome abnormalities, are the most frequent genetic triggers of POI [11]. Turner syndrome affects around 64 per 100,000 newborns [12], with over 80% of patients experiencing absent spontaneous menstruation; even among those with spontaneous menstruation, about one-third are diagnosed with POI [13]. In women carrying fragile X premutation, the incidence of POI ranges from 15–24% [14]. The genetic basis of POI is highly diverse, with various gene mutations, such as cytoplasmic polyadenylation element-binding protein 3 (CPEB3), transmembrane and coiled-coil domains 1 (TMCO1), bone morphogenetic protein-15 (BMP15), basonuclin 1 (BNC1), and others, linked to POI development [15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31]. Additionally, a recent cohort study has identified twenty more POI-associated genes involved in gonadogenesis, meiosis, follicular development, and ovulation [32]. Similarly, epigenetic modifications play a crucial role in POI pathogenesis [33, 34]. Epigenetics refers to heritable gene expression modifications that do not alter the DNA sequence [35], encompassing DNA methylation, histone modification, non-coding RNAs, and chromatin remodeling [36]. Olsen KW et al. have demonstrated that ovarian granular cells (GCs) in women with diminished ovarian reserve (DOR) exhibit distinct epigenetic features, including increased DNA methylation variability [37, 38]. What’s more, aberrant DNA methylation [39, 40], Tet1 demethylase deficiency [41], dysfunction in polycomb repressive complex 1 [42], and various non-coding RNAs, including long noncoding RNA, microRNA-127-5p, miR-379-5p, and miR-15b [43,44,45,46,47,48] may contribute to POI development by disrupting epigenetic processes. (Table 1).
Iatrogenic factors play a significant role in the etiology of POI, accounting for approximately 25% of POI cases resulting from radiotherapy, chemotherapy, and surgery [77]. Treatment with cisplatin, cyclophosphamide, or adriamycin can trigger the accelerated activation or death of primordial follicles, increase follicular atresia, damage ovarian blood vessels and stroma, ultimately leading to POI [78, 79]. It is worth noting that the impact of chemotherapeutic drugs on reproductive health varies based on the specific drug, dosage, and treatment duration [80]. Similarly, both radiotherapy, especially pelvic radiotherapy, and laparoscopic ovarian cystectomy elevate the risk of POI [81,82,83,84,85].
Autoimmune diseases such as Hashimoto's thyroiditis, Addison's disease, systemic lupus erythematosus, celiac disease, antiphospholipid syndrome, and rheumatoid arthritis contribute to approximately 4–30% of POI cases [86,87,88,89,90]. Other factors linked to POI include chronic stress, galactosemia, viral infections like mumps and HIV, and unhealthy lifestyles [91,92,93,94,95]. In addition, although controversial [96], papillomavirus vaccines are also potentially associated with an increased risk of POI [97, 98]. Additionally, environmental agents represent another significant aspect. With the ongoing global industrialization, environmental pollution has emerged as a pressing global concern. Consequently, individuals are inevitably exposed to various ETs in their daily lives. Unfortunately, the detrimental effects of ETs exposure on female reproductive health may not be fully recognized. Recent studies have highlighted the substantial role of ETs exposure in POI development [99], including atmospheric particulate matter (PM), endocrine disrupting chemicals (EDCs), pesticides, induced microplastics, heavy metals, and cigarette smoke [100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121]. Hence, a comprehensive understanding of the pathological mechanism underlying POI, particularly those by ETs exposure, could aid in the prevention and management of POI. (Fig. 1 and Table 2).
Pathological mechanisms of POI
The quantity and quality of oocytes in the ovaries, also called ovarian reserve, serve as indicators of female reproductive capacity [216]. Theoretically, women are born with a restricted number of follicles, with approximately 7 million dormant follicles present in the female fetal ovaries at around 20 weeks of gestation, representing the peak number of follicles. Over time, this number gradually lessens. At birth, the ovary typically contains one million to two million primordial follicles, which diminish to 400,000 to 500,000 by menarche. Throughout the reproductive lifespan, the majority of follicles undergo atresia, with only about 400 follicles maturing and ovulating in sequence. Menopause occurs when the number of primordial follicles in the ovary drops below 1000, triggering a series of physiological changes [217,218,219,220]. The female reproductive lifespan mostly depends on the delicate interplay between dormancy, activation and apoptosis of primordial follicles [94, 221]. Research suggests that the incidence of POI may be caused by premature primordial follicle activation, increased activation rate of primordial follicles, follicular maturation inhibition, or apoptosis acceleration [94, 222]. Therefore, protecting the quantity and quality of follicles is essential for female reproductive health. The pathological mechanisms of POI mainly involve DNA damage, oxidative stress, epigenetic modification, endocrine disorders, ovarian inflammation and ovarian cell death. (Fig. 2).
The development of follicles will go through the stages of primordial follicles, primary follicles, secondary follicles to antral follicles. the dominant follicles will be excluded and other follicles in the same period will be atresia. Various factors, including idiopathic factors, genetic factors, iatrogenic factor, autoimmune diseases, infectious diseases, environmental toxicants, can induce accelerated activation or death of primordial follicles and accelerated atretic follicles, leading to premature ovarian insufficiency.
DNA damage
DNA is the fundamental basis of genetic information, and its integrity is essential for sustaining life and maintaining health. However, the genome is inevitably under threat from various forms of DNA damage, including base damage and glycosyl destruction, base mismatch, covalent cross-linking of DNA strands, and DNA strand breaks [223]. DNA damage plays a significant role in the pathogenesis and progression of POI [224, 225]. A genetic analysis involving nearly 70,000 women identified 44 genes associated with POI, many of which are associated with DNA damage and repair processes [226, 227]. Numerous endogenous or exogenous factors can induce or aggravate DNA damage [228]. Reactive oxygen species (ROS) and reactive nitrogen are thought to be the primary endogenous chemicals responsible for DNA damage [229]. Similarly, metabolites such as acetaldehyde or S-adenosylmethionine have the similar effect [230, 231]. ROS are implicated in oxidative assaults on DNA, resulting in oxidized base fragments, oxidized sugar fragments, apurinic/apyrimidinic sites, and strand breaks [232], with DNA double-strand breaks (DSBs) being the most damaging [233]. Exogenous factors that induce DNA damage or DSBs include ionizing radiation, chemotherapy, and ETs [234,235,236,237]. For example, treatment with cyclophosphamide, adriamycin, and cisplatin can induce DSBs in primordial follicle oocytes, ultimately resulting in oocyte death [238, 239]. X-rays and gamma-rays cause DNA damage in oocytes within primordial follicles by either directly oxidizing bases or indirectly generating ROS [235]. Furthermore, various ETs exposure leads to DNA damage in oocytes, GCs, and even entire ovarian cells, including polystyrene microplastics [122,123,124,125], pesticides (such as captan, acrylonitrile, thiamethoxam, azadirachtin-based biopesticide, malathion, glyphosate, methoxychlor [110, 127,128,129,130, 132, 135]), heavy metals (for instance nickel oxide nanoparticles, silver nanoparticles, mercury chloride, strontium, aluminum, iron, cadmium, arsenic, and copper [145, 146, 148, 150, 152, 160, 240] [163]), EDCs (like di-(2-ethylhexyl) phthalate (DEHP), butylparaben, aroclor 1254, 4-vinylcyclohexene diepoxide (VCD), perfluorohexane sulfonate nonylphenol [116, 182, 183, 199, 212, 213]), cigarette smoke, dibenzo[def,p]chrysene, nicotine [170, 175, 176], and PM [241]. In daily life, exposure to one or more ETs is unavoidable, which potentially induce DNA damage through various mechanisms, including direct harm to DNA molecules, induction of DNA oxidative damage by increasing ROS levels, or interference with DNA damage repair pathways, which may overlap. For example, bisphenol A, dibutyl phthalate, perfluorooctanoic acid, DEHP, or PM not only induce DNA oxidative damage but also impact the expression of DNA damage response genes [103, 186,187,188,189, 200, 201, 205, 242, 243].
In response to DNA damage, cells activate various DNA repair pathways, including base‐excision repair, nucleotide excision repair, homologous recombination, and non-homologous end joining pathways [244]. Notably, while DNA repair mechanisms are essential for maintaining genomic stability, they produce intermediate products such as apurinic/apyrimidinic sites that can exacerbate DNA damage [245], implying that DNA damage can set off a vicious cycle of DNA damage and potentially leading to cell death. Furthermore, when DNA damage is too severe to repair or DNA repair mechanisms are overwhelmed, various cellular responses are activated, including cell cycle arrest, senescence, apoptosis, autophagy or necrosis [246,247,248], which are at least partially achieved by regulating the HUS-1-CEP-1-EGL-1-CED-9-CED-4-CED-3 pathway [122] or the PI3K/Akt pathway [138]. Turan V et al. demonstrated that the accumulation of DSBs in primordial follicles accelerates follicular apoptosis, diminishes ovarian reserve, and ultimately accelerates ovarian aging [227]. (Fig. 3).
Various factors, including DNA repair deficiency, chemotherapy, radiation, reactive oxygen species and environmental toxicants, can induce DNA damage. To response DNA damage, DNA repair pathways are triggered. The DNA repair intermediate products may exacerbate DNA damage. When DNA damage is too severe to repair or DNA repair mechanisms are overwhelmed, cell cycle arrest, senescence, apoptosis, autophagy or necrosis is triggered.
Oxidative stress
Oxidative stress is a state of imbalance between oxidation and anti-oxidation caused by the overproduction of oxidants [249]. ROS are widely recognized as prevalent oxidants, including superoxide anion, hydrogen peroxide, hydroxyl radical, ozone, and singlet oxygen [250]. Studies have shown that physiological ROS levels are critical in embryonic and follicular development, steroidogenesis, and oocyte maturation [251,252,253,254]. However, excessive ROS cause oxidative damage to DNA, proteins, lipids, organelles, and cell membranes, which may trigger cell death pathways such as apoptosis, autophagy, ferroptosis, pyroptosis, necrosis, and ultimately lead to follicular atresia and reproductive aging [115, 116, 249, 255,256,257,258].
Oxidative stress is one of the culprits of POI, with various pathogenic factors of POI leading to ovarian damage via this pathway. For example, BNC1 is important in maintaining the redox homeostasis and lipid metabolism balance of oocytes. Mutation or deficiency of BNC1 causes an increase in mitochondrial membrane potential, ROS, and mitochondrial superoxide indicator levels in oocytes, ultimately triggering oocyte ferroptosis via the NF2-YAP signal pathway [18]. CPEB3 regulates mitochondrial activity, intracellular ROS concentration, mitochondrial activity, cell proliferation, and apoptosis in cumulus cells [259], whereas CPEB3 deficiency disrupts follicular development and leads to POI [15]. In addition, deficiencies or mutations in TMCO1, BMP15, and the follicle-stimulating hormone receptor are also linked to increased ovarian ROS and oxidative stress [16, 260,261,262]. Similarly, chemotherapy induces excessive ROS production and oxidative stress in the ovaries, which leads to mitochondrial dysfunction, lipid peroxidation, ovarian cell death, and follicle loss [53, 257, 263,264,265]. Radiation exposure also caused ovarian oxidative damage by increasing ROS levels, inhibiting glutathione peroxidase activity, and elevating NADPH oxidase subunits [266,267,268]. ETs exposure is a significant contributor to ovarian oxidative stress. Studies have demonstrated that PM exposure not only induces high levels of ROS and oxidative stress in the ovary by activating the NF-κB/IL-6 signaling pathway [215, 241, 269], but also lowers the ovarian reserve of offspring mice by activating ROS-dependent NF-κB and PI3K/Akt/FoxO3a signaling pathways [270]. Similarly, microplastic exposure raises ROS levels in oocytes or GCs, causing DNA oxidation, mitochondrial dysfunction, cell cycle arrest, apoptosis, and necrosis [114,115,116, 124, 125, 271]. Furthermore, pesticides (including captan, organochlorine pesticides, acrylonitrile, thiamethoxam, organophosphorus insecticide diazinon, malathion, acetochlor, allethrin, pyriproxyfen, mancozeb, imidacloprid, glyphosate [108, 110, 127, 128, 130, 131, 133, 134, 137,138,139,140,141,142]), heavy metals (including iron overload, silver nanoparticle, cadmium, copper nanoparticle, lead, mercury, sodium arsenite [147, 149, 153,154,155, 160, 164, 166,167,168,169, 272, 273]), EDCs (including nonylphenol, VCD, bisphenol A, bisphenol S, bisphenol F, and bisphenol AF, fluorene-9-bisphenol, DEHP, butylparaben, perfluorooctanoic acid, propylparaben [116, 182, 183, 190, 191, 198, 199, 202, 203, 210, 274]), cigarette smoke, 3-monochloro-1,2-propanediol, and nicotine [170, 171, 177, 181] can induce oxidative damage in ovarian cells through different mechanisms, including increasing levels of ROS, reducing antioxidant enzyme activity, increasing oxidase activity, triggering endoplasmic reticulum stress, and impairing mitochondrial function, ultimately resulting in decreased oocyte quality, cell death, follicular atresia, and ovarian dysfunction. Paradoxically, Shi L et al. suggested that long-term moderate oxidative stress induced by ROS may only impact the quality of oocytes and follicles, but not the quantity of oocytes [275]. The contradictory conclusion could be attributed to differences in ROS levels and the specific context of the study. Notably, it is critical to recognize that oxidative stress is multifaceted, making its extent and potential hazards difficult to precisely control. (Fig. 4).
Gene mutation or deficiency, chemotherapy, radiation or environmental toxicants can decrease glutathione peroxidase activity, increase NADPH oxidase subunits activity and induce the high level of ROS, leading to lipids/proteins peroxidation, endoplasmic reticulum stress, mitochondrial dysfunction, apoptosis, autophagy, ferroptosis, pyroptosis, necrosis, which ultimately lead to follicular atresia and premature ovarian insufficiency.
Epigenetic modifications and pathophysiology of POI
Epigenetic modification regulates gene expression through DNA methylation, histone modification, and RNA interference, playing a crucial role in establishing primordial follicle pools [42], oogenesis, and embryonic development [276]. Abnormal epigenetic modifications, such as significantly increased H3K9ac and H3K9me2 modifications in the regulatory region of the FMR1 gene in GCs [277], epitope mutations in mural GCs, increased DNA methylation variability, and predicted telomere length, are common in patients with POI [37].
Various pathogenic factors of POI may induce ovarian damage through interfering with epigenetic modification. Roy et al. found that the histone demethylase JMJD3 in ovarian GCs is associated with the demethylation of H3K27, and conditional deletion of JMJD3 will lead to a decreased number of healthy follicles, estrous cycle disorders, and increased follicular atresia, eventually resulting in POI and subfertility [278]. Yang et al. demonstrated that cyclophosphamide alters DNA methylation and histone modifications in germinal vesicle oocytes, exhibited as increased H3K9me3 levels, decreased H3K4me3 levels, and 5hmC/5mC ratio, resulting in poor oocyte quality [279]. Pre-conceptional maternal exposure to cyclophosphamide can alter the methylation levels of imprinted genes (H19, Igf2r and Peg3) in offspring oocytes, impairing oocyte quality and function [280]. Furthermore, ionizing radiation alters epigenetic modifications in female germ cells, which are primarily mediated by chromatin remodeling and telomere function changes [281]. Similarly, ETs exposure causes abnormal epigenetic alteration, which contributes to the onset and progression of POI [104]. For example, bisphenol A exposure affects steroidogenesis and reproductive function by interfering with DNA methylation and histone trimethylation levels (H3K4me3, H3K27me3 and H3K9me3) in the ovary [192, 193]. Acute low-dose exposure to bisphenol S increases 5-methylcytosine and H3K27me2 in immature oocytes or 5-methylcytosine during oocyte maturation, resulting in lower oocyte quality [196]. DEHP exposure affects the expression of LIM homeobox 8 and SOHLH1 in oocytes and primordial follicle assembly via inhibiting the expression of Smyd3-H3K4me3 in ovaries [206]. Perinatal propylparaben exposure causes steroidogenesis dysfunction and ovarian aging in offspring mice, which may be linked to Cyp1a1 hypermethylation and changes in Peg3 and H19 methylation levels [211]. According to Li et al., ovarian damage and POI produced by VCD exposure were associated with lower levels of N6-methyladenosine, its regulator demethylase ALKBH5, and ALKBH5-mediated YAP activity [184]. Low-dose chlordecone exposure during pregnancy decreases ovarian reserve and increases follicular atresia in female offspring, which may be attributed to an increase in H2Aub and H3K27me3, a decrease in H4ac and H3K4me3 in embryonic oocytes, and a decrease in histone H3 trimethylation and H4 acetylation in growing oocytes [143]. Furthermore, heavy metal cadmium impairs follicular development, steroidogenesis, and GCs function by interfering with DNA methylation, miRNAs (miR-27a-3p, miR-27b-3p, miR-146, miR-10b-5p, and miR-211-3p), or m6A [156,157,158]. Long-term arsenic exposure causes genotoxicity and reproductive harm by reducing DNA methylation levels in the ovary [161]. Meanwhile, arsenic exposure raises the level of DNA methylation in the promoter region of steroidogenic factor-1 in GCs, resulting in aberrant steroidogenesis [162]. Cigarette smoke exposure can alter the methylation of interleukin-6 gene in cumulus cells, potentially affecting oocyte maturation [171]. Furthermore, prenatal nicotine exposure promotes ovarian dysplasia and interferes with estradiol synthesis in offspring rats, which may be associated with reduced H3K27ac and H3K9ac levels in the promotor region of the cytochrome P450 aromatase via nicotinic acetylcholine receptors [178].
Endocrine disruption, altered steroidogenesis and POI
Estradiol, androgens, FSH, and anti-Müllerian hormone, as well as their interactions, play a crucial role in folliculogenesis and the maintenance of ovarian reserve [95]. Consequently, disorders in their synthesis and secretion potentially impact ovarian reserve. EDCs are a type of natural or synthetic compounds that interferes or disrupts the normal endocrine function. Numerous studies have confirmed that EDCs exposure is a significant environmental risk factor and determinant of POI [282]. These substances disrupt the endocrine system by interfering with hormone synthesis or receptor expression, altering circulating hormone levels or distribution, interacting with hormone receptors, affecting signal transduction, inducing epigenetic modification, and altering hormone metabolism or clearance [283]. For example, bisphenol A exhibits a strong affinity for estrogen-related receptor-γ and G protein-coupled receptor-30, potentially disrupting the interaction between estradiol and its receptors [194]. Polycyclic aromatic hydrocarbons not only interact with estradiol receptor α and G protein-coupled receptors to inhibit estradiol production in GCs but also induce the expression of Bax in oocytes through the aryl hydrocarbon receptor, ultimately leading to apoptosis [214]. Hao et al. indicated that exposure to chlorothalonil reduces the level of estrogen receptor α in the ovary, potentially resulting in endocrine disorders [144]. Thiamethoxam exposure disrupts the levels of female hormone receptors, including anti-Müllerian hormone receptor, estrogen receptor β, follicle-stimulating hormone receptor, and luteinizing hormone receptor in mouse ovaries [128]. Furthermore, substances like phthalates, parabens, perfluorooctanoic acid/perfluorooctane sulphonic acid, diethylstilbestrol, 2,3,7,8-tetrachlorodibenzo-p-dioxin, triclosan can interfere with ovarian steroidogenesis and function [104, 204, 207]. Interestingly, EDCs can also impact non-estrogen pathways by interfering with the expression of genes relevant to primordial germ cell development and affecting oocyte meiosis to impair follicular development [284, 285]. Cigarette smoking causes endocrine disorders by reducing estrogen expression, accelerating estrogen clearance, and increasing serum androgen levels to antagonize estrogen [95]. PM2.5 exposure disrupts ovarian endocrine function, as evidenced by a significant decrease in estradiol and steroid synthase (including Cyp11a, Cyp19a, Cyp17a, and Hsd3b) and a considerable increase in FSH [215]. Furthermore, cadmium exposure alters hormone synthesis function in offspring GCs by modulating miRNAs and DNA methylation modifications [159].
Ovarian inflammation and POI
Animal studies have demonstrated a significantly increase in pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, and iNOS) in the ovaries of POI models, with anti-inflammatory and anti-oxidative stress treatments showing efficacy in improving ovarian function [82, 286, 287]. Meanwhile, clinical studies have indicated that helper T cell 1 levels in POI females'peripheral blood and ovaries are significantly higher [288], and the cell-mediated immune response is positively associated with inflammatory response [289]. In addition, an increasing number of studies have demonstrated that inflammatory aging, a chronic low-grade inflammation status with aging, plays an important role in the pathogenesis of POI [289, 290], confirming the strong association between ovarian inflammation and POI. It is possible that any factor contributing to ovarian inflammation could lead to POI.
Autoimmune diseases are one of the most important pathogenic factors in POI, since they induce inflammatory autoimmune responses in tissues and increase pro-inflammatory signals [291, 292]. It is plausible to suggest that POI induced by autoimmune diseases is largely connected to ovarian inflammation. Al-Shahat et al. indicated that cisplatin induces ovarian inflammation by activating the p38/MAPK and JNK signaling pathways, as evidenced by the upregulation of pro-inflammatory markers (IL1β, IL6, TNF-α, NF-κβ, and TGF-β1) and the downregulation of anti-inflammatory markers (IL10), ultimately leading to ovarian dysfunction [293]. Moreover, the binding of TNF-α to its receptor activates MAPKs and NF-κB pathways, initiating an inflammatory signal amplification loop that leads to more severe tissue damage [294]. Similarly, cyclophosphamide or radiotherapy treatment can induce ovarian inflammation and upregulate the expression of pro-inflammatory factor (TNF-α) [78, 295, 296]. High levels of TNF-α may promote intracellular calcium accumulation and activate endonucleases, ultimately inducing apoptosis of growing follicles [297]. What’s more, ovarian inflammation induced by ETs contributes to the development POI. For example, PM2.5 triggers ovarian oxidative stress and inflammation by activating the NF-κB/IL-6 signaling pathway, leading to ovarian dysfunction and decreased ovarian reserve [215]. Concurrently, it promotes GCs apoptosis and follicular atresia by inducing an inflammatory response [102]. Saeed et al. showed that microplastic-induced ovarian damage is linked to increased levels of pro-inflammatory factors (IL-6 and NF-κB) [126]. Exposure to bisphenol S can induce an ovarian inflammatory response via SIRT-1/Nrf2/NF-κB pathway, leading to increased levels of inflammatory mediators such as TNF-α, CRP, iNOS, and COX-2 [197]. Additionally, perinatal BPA exposure can impair female offspring's reproductive function, possibly through inflammatory response and abnormal autophagy mediated by the TLR4/NF-κB and mTOR signaling pathways [195]. DEHP induces inflammation-related cell death (pyroptosis) in GCs via SLC39A5/NF-κB/NLRP3 pathway [208]. Meanwhile, DEHP-induced ovarian inflammation is associated with intestinal flora imbalances and alterations in fecal metabolites [209]. Moreover, ovarian dysfunction caused by captan [110], thiamethoxam [128], organochlorine pesticides [137], and 3-monochloro-1,2-propanediol [181] is also associated with the induction of ovarian inflammation.
Ovarian cell death and POI
Throughout the reproductive lifespan, regulated cell death plays a vital role in follicular formation, follicular development, follicular atresia, dominant follicle excretion, and removal of damaged oocytes. However, endogenous or exogenous factors may trigger the regulated cell death pathway and lead to follicular loss [298], which may be closely associated with the occurrence and progression of POI.
Apoptosis is the most extensively studied pathway of regulatory cell death in the ovary [299]. It is primarily triggered by two pathways: the mitochondrial pathway and the death receptor pathway. The mitochondrial pathway is regulated by members of the B-cell lymphoma 2 protein family, while the death receptor pathway is mainly initiated by extracellular ligands that bind to death receptors and trigger apoptosis [298, 300, 301]. However, it is worth noting that the two pathways are not completely independent [301]. Several studies have shown that gene deletion or abnormality (including CPEB3, TMCO1, BMP15), ionizing radiation, chemotherapy, and ETs cause ovarian dysfunction and POI by inducing apoptosis of ovarian somatic cells or oocytes [15, 16, 79, 191, 267, 270, 302, 303].
In addition to apoptosis, ferroptosis, autophagy and autophagic cell death, pyroptosis, necrosis, and necroptosis also play an important role in the development of POI [304,305,306,307,308,309]]. For example, BNC1 deficiency induces oocyte ferroptosis through the NF2-YAP pathway [18]. Cisplatin induces ferroptosis in GCs by regulating the expression of ferroptosis-related molecules, including glutathione peroxidase 4, acetyl CoA synthetase long chain family member 4, arachidonic acid 15-lipoxygenase, and solute carrier family 7 member 11, leading to follicular development disorders and POI [304]. Delcour et al. found that mutations in autophagy-related genes ATG7 and ATG9A reduce autophagosome synthesis and impair autophagy, which is closely related to decreased ovarian reserve [310]. In addition, Liu et al. showed that Tet1 deficiency causes a declined in autophagy and ubiquitination, elevating organelle fission in oocytes [41]. VCD triggers autophagy to induce POI after inactivating the Akt/mTOR pathway by inhibiting miR-144 [185]. In addition, abnormalities in autophagy caused by cyclophosphamide, tripterygium glycosides, diazinon, malathion, and acrylonitrile are closely associated with ovarian dysfunction [48, 127, 131, 138, 311, 312]. Meanwhile, cyclophosphamide or microplastics exposure can also cause POI by inducing pyroptosis, which may be related to the NLRP3/Caspase-1 signaling pathway [115, 306, 313].
Treatment strategies
In recent years, considerable efforts have been devoted to exploring effective treatments for POI. Unfortunately, no entirely effective and optimal treatment has yet been identified. Current therapeutic approaches for POI include hormone replacement therapy (HRT), stem cell and exosome therapy, melatonin therapy, traditional Chinese medicine therapy, in vitro activation, platelet-rich plasma (PRP) therapy, and ovarian tissue cryopreservation. (Fig. 5).
Hormone replacement therapy
HRT, the predominant clinical intervention for POI, aims to raise sex hormone levels to premenopausal levels through exogenous supplementation. This approach effectively alleviates symptoms associated with POI and lowers the risk of chronic diseases such as osteoporosis and cardiovascular disease [314,315,316]. Regrettably, HRT treatment does not fundamentally restore ovarian reserve in patients, and long-term HRT treatment may even increase the risk of breast cancer and venous thrombosis [317, 318]. Conversely, there isn't any solid proof yet that HRT increases the risk of cancer recurrence in those who have survived cancer [319, 320]. Therefore, HRT can be administered to POI patients without a contraindication to alleviate menopausal symptoms and mitigate the risk of disease stemming from hormone deficiency.
Stem cell and exosome therapy
Stem cells are undifferentiated cells that have the potential to multi-differentiate, proliferate indefinitely, and self-renew. According to the source, stem cells are classified as adult stem cells, induced pluripotent stem cells, and embryonic stem cells [321]. Mesenchymal stem cells (MSCs), a type of adult stem cells, are the most extensively investigated in POI stem cell therapy [289, 322]. MSCs are extensively derived from bone marrow, fat, umbilical cord, amniotic membrane, menstrual blood, and embryos, and they benefit from low immunogenicity and easy access [323]. Animal studies have confirmed that MSCs transplantation effectively restores the ovarian function of the POI model [324,325,326,327]. Its mechanism may be related to homing, differentiation, paracrine, angiogenesis, immune regulation, anti-inflammatory, anti-apoptosis, anti-fibrosis, and anti-oxidative stress [322, 323, 328]. Meanwhile, clinical studies have been conducted gradually [323]. However, while MSCs have a strong therapeutic potential in POI, their safety has aroused concerns. First, MSCs production and preparation have not yet been standardized, which may result in uneven quality of cells; second, there is still a lack of standards to characterize the safety, biological distribution, and effectiveness of MSCs, making it difficult to evaluate the therapeutic effect; and finally, MSCs therapy may have potential risks of tumorigenicity, immune rejection, and chromosomal aberrations [329,330,331]. To prevent the potential hazards of MSCs treatment, exosome therapy is being investigated progressively.
Exosomes are nano-sized vesicles secreted by cells that contain proteins, lipids, nucleic acids, and other substances [331]. Similar to MSCs, exosomes improve ovarian function in POI animals by inhibiting apoptosis, promoting angiogenesis, reducing oxidative stress, preventing fibrosis, and promoting cell proliferation, implying that exosomes represent a promising cell-free treatment for POI. However, it is worth noting that the generation of exosomes requires standardized production and purifying techniques. Meanwhile, the disadvantages of exosome therapy, such as low targeting, low retention, and immunogenicity, require improved solutions [332].
Melatonin treatment
Melatonin was originally assumed to be an amine hormone secreted by the pineal gland [333]. Recent studies have shown that it can also be synthesized in other tissues such as ovary, testis, placenta, and skin [305]. Melatonin has a beneficial effect in POI, reducing cyclophosphamide-induced primordial follicle loss and inhibiting GC apoptosis by regulating the PTEN/Akt/FoxO3a pathway [334, 335]. Meanwhile, it regulates the ERK pathway, which protects ovarian cells from excessive autophagy-induced ovarian reserve loss and mitochondrial dysfunction [305]. In addition, the DNA protective properties and antioxidant activities of melatonin aid to protect ovarian function and fertility in POI patients [336]. In summary, melatonin supplementation is thought to be a treatment for improving ovarian function in women with POI.
Traditional Chinese medicine treatment
Traditional Chinese medicine has a definite effect in the treatment of POI [337,338,339,340]. The mechanism includes anti-apoptosis [341,342,343], antioxidative stress [344], immunological regulation [337], angiogenesis [345], autophagy regulation [346], DNA damage repair [347], and epigenetic modification [348, 349]. Si-Wu-tang therapy improves follicular development, ovarian function, antioxidant capacity, and angiogenesis in POI mice, possibly through regulating the PI3K/Akt, Nrf2/HO-1, and STAT3/HIF-1α/VEGF signaling pathways [345, 350]. Jian-Pi-Yi-Shen decoction improves mitochondrial activity by regulating the ASK1/JNK signaling pathway, which inhibits GC apoptosis [342]. Similarly, Huyang yangkun formula promote follicular development by regulating AQP8/Bcl-2 family-related apoptosis [341], as well as the Hippo/JAK2/STAT3 pathway to improve ovarian function in POI rats [351]. Zigui-Yichong-Fang is involved in mediating Foxo3 a deacetylation by regulating SIRT1/FoxO3a pathway, which improvs ovarian reserve in POI mice [348]. Bushen-Culuan Decoction may improve ovarian oxidative stress and ovarian reserve by activating the Nrf2/ARE signaling pathway [352]. Similarly, Bu Shen Huo Xue Tang effectively protects the ovarian function of autoimmune POI mice by activating the Nrf2/Keap1 pathway and enhancing antioxidant capacity [344]. Furthermore, it protected ovarian reserve and function in POI animals, maybe connected with promoting endogenous BMSC proliferation and homing [353]. Clinical studies have also shown that Zishen Yutai Pills has a positive effect on patients with DOR undergoing in vitro fertilization and embryo transfer, as evidenced by an increase in the number of oocytes and embryos [354]. Traditional Chinese medicine monomers, such as resveratrol, quercetin, (-)-epicatechin, polysaccharides, honokiol, lycium barbarum polysaccharide, red ginseng, icariin, ginsenoside Rg1, and chrysin, play an excellent role in POI treatment [286, 287, 306, 347, 355,356,357,358,359,360,361,362,363].
Although Chinese herbal compounds have great therapeutic potential in POI, their safety has always been a concern due to the potential toxic components in Chinese herbal medicines, such as aristolochic acid, aconitine, triptolide, brucine, aconite, and metals [364,365,366], which severely limit the global development of traditional Chinese medicine [367, 368]. As a result, how to capitalize on the benefits of traditional Chinese medicine therapy and advance toward internationalization remains a huge challenge for the development of traditional Chinese medicine.
Acupuncture is more generally acknowledged than Chinese herbal medicine treatment. It not only effectively improves the hormone disorder in POI model and inhibits GC apoptosis [369,370,371,372], but it also protects ovarian function by regulating intestinal microflora alterations and inhibiting ovarian oxidative stress and Fe2+ accumulation [373]. Similarly, clinical studies have shown that acupuncture has a considerable effect on inhibiting apoptosis, increasing the number of antral follicles, and improving sex hormone levels and ovarian function in patients with DOR or POI [374]. In addition, moxibustion can effectively protect the ovarian function of POI models or patients [375, 376], which may be achieved by inhibiting PI3K/Akt/mTOR signaling pathway [375] or mitigating NLRP3 inflammatory activation and mitochondrial dysfunction [377].
In vitro activation
In vitro activation (IVA) is a technique that stimulates the growth of residual primordial follicles and promotes their development into functional oocytes, hence improving the oocyte retrieval rate of POI patients [378, 379]. The activation of PTEN/PI3K/Akt/Foxo3 signaling pathway, as well as the inhibition of the Hippo signaling pathway, play critical roles in IVA-induced follicular activation and development [380, 381]. Clinical studies have also confirmed that drug-free IVA combined with laparoscopic ovarian incision treatment can disrupt the Hippo signaling pathway and promote the growth of residual follicles in the ovaries of POI patients [382, 383]. In addition, ovarian biopsy and ovarian scraping techniques can promote follicular development in vivo [384]. Interestingly, Lunding et al. found that, while more than half of POI patients became pregnant after undergoing ovarian biopsying, fragmentation, and autotransplantation, there is no conclusive evidence that this treatment increases the number of retrieved oocytes for IVF/ICSI after 10 weeks [385]. In addition, while Zhai et al. showed that no side effects from the IVA operation after 2–4 years of follow-up [379], the long-term implications of IVA procedure remained unknown. Therefore, the safety and efficacy of IVA treatment still need further exploration.
Platelet-rich plasma therapy
PRP is a biological product derived from autologous blood that contains various growth factors, cytokines, proteins, and hormones [386]. Studies have shown that intraovarian PRP injection may be an effective strategy for treating POI. It can improve sex hormone levels, restore the menstrual cycle, promote primordial follicle development, and increase oocyte retrieval rates, all while improving the reproductive potential of POI patients [386,387,388]. However, the mechanism of PRP, particularly in POI, remains mostly unexplained. Ahmadian et al. showed that PRP stimulated ovarian angiogenesis while inhibiting follicular degeneration and atresia induced by ovotoxic substances. Meanwhile, it has considerable potential to inhibit inflammation, restore tissue damage, and stimulate tissue regeneration [389]. In addition, the protective effect of PRP may be linked to apoptosis inhibition and mTOR signaling activation [390]. Although PRP has great potential for treating POI, there are also potential risks, including infection, tumorigenesis, immunological rejection, and embryo safety [380, 391, 392]. Moreover, the preparation technique, quality evaluation, dosage, and duration of PRP have yet to be standardized, limiting its use significantly. Therefore, future research still needs to further clarify the molecular mechanism of PRP in the treatment of POI, as well as develop a standardized process for PRP preparation and administration.
Other treatments
Ovarian tissue cryopreservation and autologous transplantation effectively restore the reproductive endocrine function and fertility in POI patients [393, 394]. In addition, coenzyme Q10, tissue engineering techniques are also expected to protect the ovarian function in POI patients [395, 396]. Meanwhile, it is also crucial to keep a healthy lifestyle [95, 397].
Discussion
POI is a complex gynecological endocrine disease, and its etiology is highly heterogeneous, including genetic defects, epigenetic changes, iatrogenic injuries, autoimmune diseases, and environmental factors [2]. They may cause ovarian dysfunction and ovarian reserve depletion by inducing DNA damage and oxidative stress, changing epigenetic modification, disrupting endocrine function, causing ovarian inflammation and ovarian cell death. Nowadays, although people are gradually aware of the potential harm of environmental factors on ovarian function, it is far from enough compared with genetic defects and iatrogenic factors [78, 398,399,400]. The reasons may be as follows: firstly, ovarian damage induced by ETs may be caused by prolonged and continuous exposure, while current studies mostly focus on a single time point, which may lead to the inability to identify ovotoxic substances in time; secondly, the diagnosis of POI is mostly concentrated in women aged 30–40 years old, but the time point of ETs exposure may have occurred many years ago, which also makes it difficult for us to find the exact cause; thirdly, The complexity and unknown of POI pathological mechanism and oogenesis process. Therefore, more prospective and retrospective cohort studies and experimental studies are needed to find the ovotoxic ETs and their effects on ovarian function, as well as elucidate the related molecular mechanism.
However, in the face of known ETs, how to avoid the potential harm remains a difficult problem. As we all know, there are many kinds of ETs, and they are everywhere in our lives. For example, bisphenol A is widely used in the manufacture of polycarbonate, acrylate and epoxy resin [401, 402]. Phthalate is often used in the manufacture of toys and food packaging materials [403, 404]. Parabens are widely found in preservatives in pharmaceuticals, food, industrial products and personal care products [405]. Plastic products are widely used in packaging, construction, automobile, furniture, agriculture and cosmetics [406]. Furthermore, pesticide is an indispensable part of the development of agriculture, forestry and animal husbandry [407]. These mean that it is very difficult to completely avoid these ovotoxic substances. Therefore, it is urgent to reduce the production, use and distribution of these ETs, as well as to research and development safer and more effective alternatives. Meanwhile, people should also consciously stay away from harmful ETs. Last but not least, ETs exposure not only impairs reproductive health [284, 408, 409], but also affects the health of offspring through changing epigenetic modification [410, 411]. Therefore, it is necessary to study the genetic variation of exposed populations in the future.
Conclusion
In a word, this paper reviews the risk factors, pathogenesis and treatment strategies of POI. Meanwhile, various ovotoxic ETs and their mechanism leading to ovarian reserve decline and ovarian dysfunction are described in detail. It is hoped to provide better strategies for the treatment and prevention of POI, and to make people aware of the potential harm of ETs exposure to female reproductive health.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- BMP15:
-
Bone morphogenetic protein-15
- BNC1:
-
Basonuclin 1
- CPEB3:
-
Cytoplasmic polyadenylation element-binding protein 3
- DEHP:
-
Di-(2-ethylhexyl) phthalate
- DOR:
-
Diminished ovarian reserve
- DSBs:
-
DNA double-strand breaks
- EDCs:
-
Endocrine disrupting chemicals
- ETs:
-
Environmental toxicants
- GCs:
-
Granulosa cells
- HRT:
-
Hormone replacement therapy
- IVA:
-
In vitro activation
- MSCs:
-
Mesenchymal stem cells
- PM:
-
Particulate matter
- POI:
-
Premature ovarian insufficiency
- RPR:
-
Platelet-rich plasma
- ROS:
-
Reactive oxygen species
- TMCO1:
-
Transmembrane and coiled-coil domains 1
- VCD:
-
4-vinylcyclohexene diepoxide
References
European Society for Human R, Embryology Guideline Group on POI, Webber L, Davies M, Anderson R, Bartlett J, et al. ESHRE Guideline: management of women with premature ovarian insufficiency. Hum Reprod. 2016;31(5):926–37.
Huang C, Zhao S, Yang Y, Guo T, Ke H, Mi X, et al. TP63 gain-of-function mutations cause premature ovarian insufficiency by inducing oocyte apoptosis. J Clin Invest. 2023;133(5).
Golezar S, Ramezani Tehrani F, Khazaei S, Ebadi A, Keshavarz Z. The global prevalence of primary ovarian insufficiency and early menopause: a meta-analysis. Climacteric. 2019;22(4):403–11.
Jones AR, Goh M, Langham R, Boyle J, Milat F, Ebeling PR, et al. Osteoporosis and premature ovarian insufficiency: geographic variation in clinicians’ and consumers’ knowledge gaps and barriers to care. Arch Osteoporos. 2020;15(1):38.
Stevenson JC, Collins P, Hamoda H, Lambrinoudaki I, Maas A, Maclaran K, et al. Cardiometabolic health in premature ovarian insufficiency. Climacteric. 2021;24(5):474–80.
Ates S, Aydin S, Ozcan P, Bakar RZ, Cetin C. Sleep, depression, anxiety and fatigue in women with premature ovarian insufficiency. J Psychosom Obstet Gynaecol. 2022;43(4):482–7.
Anagnostis P, Christou K, Artzouchaltzi AM, Gkekas NK, Kosmidou N, Siolos P, et al. Early menopause and premature ovarian insufficiency are associated with increased risk of type 2 diabetes: a systematic review and meta-analysis. Eur J Endocrinol. 2019;180(1):41–50.
Pirtea P, Ayoubi JM. Diminished ovarian reserve and poor response to stimulation are not reliable markers for oocyte quality in young patients. Fertil Steril. 2020;114(1):67–8.
Fraison E, Crawford G, Casper G, Harris V, Ledger W. Pregnancy following diagnosis of premature ovarian insufficiency: a systematic review. Reprod Biomed Online. 2019;39(3):467–76.
Qin Y, Jiao X, Simpson JL, Chen ZJ. Genetics of primary ovarian insufficiency: new developments and opportunities. Hum Reprod Update. 2015;21(6):787–808.
Silven H, Savukoski SM, Pesonen P, Pukkala E, Ojaniemi M, Gissler M, et al. Association of genetic disorders and congenital malformations with premature ovarian insufficiency: a nationwide register-based study. Hum Reprod. 2023;38(6):1224–30.
Gravholt CH, Viuff M, Just J, Sandahl K, Brun S, van der Velden J, et al. The Changing Face of Turner Syndrome. Endocr Rev. 2023;44(1):33–69.
Komura N, Mabuchi S, Sawada K, Nishio Y, Kimura T, Komura H. Subsequent menstrual disorder after spontaneous menarche in Turner syndrome. Clin Endocrinol (Oxf). 2021;95(1):163–8.
Allen EG, Charen K, Hipp HS, Shubeck L, Amin A, He W, et al. Refining the risk for fragile X-associated primary ovarian insufficiency (FXPOI) by FMR1 CGG repeat size. Genet Med. 2021;23(9):1648–55.
E F, Zhang H, Yin W, Wang C, Liu Y, Li Y, et al. CPEB3 deficiency in mice affect ovarian follicle development and causes premature ovarian insufficiency. Cell Death Dis. 2021;13(1):21.
Sun Z, Zhang H, Wang X, Wang QC, Zhang C, Wang JQ, et al. TMCO1 is essential for ovarian follicle development by regulating ER Ca(2+) store of granulosa cells. Cell Death Differ. 2018;25(9):1686–701.
Santos M, Cordts EB, Peluso C, Dornas M, Neto FHV, Bianco B, et al. Association of BMP15 and GDF9 variants to premature ovarian insufficiency. J Assist Reprod Genet. 2019;36(10):2163–9.
Wang F, Liu Y, Ni F, Jin J, Wu Y, Huang Y, et al. BNC1 deficiency-triggered ferroptosis through the NF2-YAP pathway induces primary ovarian insufficiency. Nat Commun. 2022;13(1):5871.
Luo W, Ke H, Tang S, Jiao X, Li Z, Zhao S, et al. Next-generation sequencing of 500 POI patients identified novel responsible monogenic and oligogenic variants. J Ovarian Res. 2023;16(1):39.
Jaillard S, McElreavy K, Robevska G, Akloul L, Ghieh F, Sreenivasan R, et al. STAG3 homozygous missense variant causes primary ovarian insufficiency and male non-obstructive azoospermia. Mol Hum Reprod. 2020;26(9):665–77.
Liu H, Guo T, Gong Z, Yu Y, Zhang Y, Zhao S, et al. Novel FSHR mutations in Han Chinese women with sporadic premature ovarian insufficiency. Mol Cell Endocrinol. 2019;492: 110446.
Yang Y, Guo T, Liu R, Ke H, Xu W, Zhao S, et al. FANCL gene mutations in premature ovarian insufficiency. Hum Mutat. 2020;41(5):1033–41.
Zhang YX, Li HY, He WB, Tu C, Du J, Li W, et al. XRCC2 mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans. Clin Genet. 2019;95(3):442–3.
He WB, Tu CF, Liu Q, Meng LL, Yuan SM, Luo AX, et al. DMC1 mutation that causes human non-obstructive azoospermia and premature ovarian insufficiency identified by whole-exome sequencing. J Med Genet. 2018;55(3):198–204.
Jaillard S, Sreenivasan R, Beaumont M, Robevska G, Dubourg C, Knarston IM, et al. Analysis of NR5A1 in 142 patients with premature ovarian insufficiency, diminished ovarian reserve, or unexplained infertility. Maturitas. 2020;131:78–86.
Bouali N, Francou B, Bouligand J, Imanci D, Dimassi S, Tosca L, et al. New MCM8 mutation associated with premature ovarian insufficiency and chromosomal instability in a highly consanguineous Tunisian family. Fertil Steril. 2017;108(4):694–702.
Potorac I, Laterre M, Malaise O, Nechifor V, Fasquelle C, Colleye O, et al. The Role of MCM9 in the Etiology of Sertoli Cell-Only Syndrome and Premature Ovarian Insufficiency. J Clin Med. 2023;12(3).
Bouali N, Francou B, Bouligand J, Lakhal B, Malek I, Kammoun M, et al. NOBOX is a strong autosomal candidate gene in Tunisian patients with primary ovarian insufficiency. Clin Genet. 2016;89(5):608–13.
D’Ignazio L, Michel M, Beyer M, Thompson K, Forabosco A, Schlessinger D, et al. Lhx8 ablation leads to massive autophagy of mouse oocytes associated with DNA damage. Biol Reprod. 2018;98(4):532–42.
Zhe J, Chen S, Chen X, Liu Y, Li Y, Zhou X, et al. A novel heterozygous splice-altering mutation in HFM1 may be a cause of premature ovarian insufficiency. J Ovarian Res. 2019;12(1):61.
Zhang Q, Zhang W, Wu X, Ke H, Qin Y, Zhao S, et al. Homozygous missense variant in MEIOSIN causes premature ovarian insufficiency. Hum Reprod. 2023;38(Supplement_2):ii47-ii56.
Ke H, Tang S, Guo T, Hou D, Jiao X, Li S, et al. Landscape of pathogenic mutations in premature ovarian insufficiency. Nat Med. 2023;29(2):483–92.
Wang J, Sun X, Yang Z, Li S, Wang Y, Ren R, et al. Epigenetic regulation in premature ovarian failure: A literature review. Front Physiol. 2022;13: 998424.
Zhang JH, Chen JH, Guo B, Fang Y, Xu ZY, Zhan L, et al. Recent Insights into Noncoding RNAs in Primary Ovarian Insufficiency: Focus on Mechanisms and Treatments. J Clin Endocrinol Metab. 2023;108(8):1898–908.
Piccinini A, Bailo P, Barbara G, Miozzo M, Tabano S, Colapietro P, et al. Violence against Women and Stress-Related Disorders: Seeking for Associated Epigenetic Signatures, a Pilot Study. Healthcare (Basel). 2023;11(2).
Yamamuro C, Zhu JK, Yang Z. Epigenetic Modifications and Plant Hormone Action. Mol Plant. 2016;9(1):57–70.
Olsen KW, Castillo-Fernandez J, Chan AC, la Cour FN, Zedeler A, Bungum M, et al. Identification of a unique epigenetic profile in women with diminished ovarian reserve. Fertil Steril. 2021;115(3):732–41.
Olsen KW, Castillo-Fernandez J, Zedeler A, Freiesleben NC, Bungum M, Chan AC, et al. A distinctive epigenetic ageing profile in human granulosa cells. Hum Reprod. 2020;35(6):1332–45.
Liu X, Nie ZW, Gao YY, Chen L, Yin SY, Zhang X, et al. Sodium fluoride disturbs DNA methylation of NNAT and declines oocyte quality by impairing glucose transport in porcine oocytes. Environ Mol Mutagen. 2018;59(3):223–33.
Kordowitzki P, Haghani A, Zoller JA, Li CZ, Raj K, Spangler ML, et al. Epigenetic clock and methylation study of oocytes from a bovine model of reproductive aging. Aging Cell. 2021;20(5): e13349.
Liu L, Wang H, Xu GL, Liu L. Tet1 Deficiency Leads to Premature Ovarian Failure. Front Cell Dev Biol. 2021;9: 644135.
Hu M, Yeh YH, Munakata Y, Abe H, Sakashita A, Maezawa S, et al. PRC1-mediated epigenetic programming is required to generate the ovarian reserve. Nat Commun. 2022;13(1):4510.
Wang X, Zhang X, Dang Y, Li D, Lu G, Chan WY, et al. Long noncoding RNA HCP5 participates in premature ovarian insufficiency by transcriptionally regulating MSH5 and DNA damage repair via YB1. Nucleic Acids Res. 2020;48(8):4480–91.
Wang F, Chen X, Sun B, Ma Y, Niu W, Zhai J, et al. Hypermethylation-mediated downregulation of lncRNA PVT1 promotes granulosa cell apoptosis in premature ovarian insufficiency via interacting with Foxo3a. J Cell Physiol. 2021;236(7):5162–75.
Li D, Wang X, Dang Y, Zhang X, Zhao S, Lu G, et al. lncRNA GCAT1 is involved in premature ovarian insufficiency by regulating p27 translation in GCs via competitive binding to PTBP1. Mol Ther Nucleic Acids. 2021;23:132–41.
Zhang X, Dang Y, Liu R, Zhao S, Ma J, Qin Y. MicroRNA-127-5p impairs function of granulosa cells via HMGB2 gene in premature ovarian insufficiency. J Cell Physiol. 2020;235(11):8826–38.
Dang Y, Wang X, Hao Y, Zhang X, Zhao S, Ma J, et al. MicroRNA-379-5p is associate with biochemical premature ovarian insufficiency through PARP1 and XRCC6. Cell Death Dis. 2018;9(2):106.
Liu T, Liu Y, Huang Y, Chen J, Yu Z, Chen C, et al. miR-15b induces premature ovarian failure in mice via inhibition of alpha-Klotho expression in ovarian granulosa cells. Free Radic Biol Med. 2019;141:383–92.
Alvaro Mercadal B, Imbert R, Demeestere I, Gervy C, De Leener A, Englert Y, et al. AMH mutations with reduced in vitro bioactivity are related to premature ovarian insufficiency. Hum Reprod. 2015;30(5):1196–202.
Li L, Zhou X, Wang X, Wang J, Zhang W, Wang B, et al. A dominant negative mutation at the ATP binding domain of AMHR2 is associated with a defective anti-Mullerian hormone signaling pathway. Mol Hum Reprod. 2016;22(9):669–78.
Liu H, Wei X, Sha Y, Liu W, Gao H, Lin J, et al. Whole-exome sequencing in patients with premature ovarian insufficiency: early detection and early intervention. J Ovarian Res. 2020;13(1):114.
Renault L, Patino LC, Magnin F, Delemer B, Young J, Laissue P, et al. BMPR1A and BMPR1B Missense Mutations Cause Primary Ovarian Insufficiency. J Clin Endocrinol Metab. 2020;105(4).
Li G, Gu J, Zhou X, Wu T, Li X, Hua R, et al. Mitochondrial stress response gene Clpp deficiency impairs oocyte competence and deteriorate cyclophosphamide-induced ovarian damage in young mice. Front Endocrinol (Lausanne). 2023;14:1122012.
Fan S, Jiao Y, Khan R, Jiang X, Javed AR, Ali A, et al. Homozygous mutations in C14orf39/SIX6OS1 cause non-obstructive azoospermia and premature ovarian insufficiency in humans. Am J Hum Genet. 2021;108(2):324–36.
Qin Y, Guo T, Li G, Tang TS, Zhao S, Jiao X, et al. CSB-PGBD3 Mutations Cause Premature Ovarian Failure. PLoS Genet. 2015;11(7): e1005419.
Kim S, Pyun JA, Cha DH, Ko JJ, Kwack K. Epistasis between FSHR and CYP19A1 polymorphisms is associated with premature ovarian failure. Fertil Steril. 2011;95(8):2585–8.
Zhao M, Feng F, Chu C, Yue W, Li L. A novel EIF4ENIF1 mutation associated with a diminished ovarian reserve and premature ovarian insufficiency identified by whole-exome sequencing. J Ovarian Res. 2019;12(1):119.
Kuang L, Liu B, Xi D, Gao Y. A novel heterozygous ERCC6 variant identified in a Chinese family with non-syndromic primary ovarian insufficiency. Mol Genet Genomic Med. 2022;10(10): e2040.
Sadat Eshaghi F, Dehghan Tezerjani M, Ghasemi N, Dehghani M. Association study of ESR1 rs9340799, rs2234693, and MMP2 rs243865 variants in Iranian women with premature ovarian insufficiency: A case-control study. Int J Reprod Biomed. 2022;20(10):841–50.
Yang X, Zhang X, Jiao J, Zhang F, Pan Y, Wang Q, et al. Rare variants in FANCA induce premature ovarian insufficiency. Hum Genet. 2019;138(11–12):1227–36.
Mei L, Huang Y, Wu X, He H, Ye R, Ma J, et al. Mutations in FIGLA Associated With Premature Ovarian Insufficiency in a Chinese Population. Front Med (Lausanne). 2021;8: 714306.
Li J, Peng T, Wang L, Long P, Quan R, Tan H, et al. Heterozygous FMN2 missense variant found in a family case of premature ovarian insufficiency. J Ovarian Res. 2022;15(1):31.
Tang R, Yu Q. The significance of FMR1 CGG repeats in Chinese women with premature ovarian insufficiency and diminished ovarian reserve. Reprod Biol Endocrinol. 2020;18(1):82.
Wang B, Mu Y, Ni F, Zhou S, Wang J, Cao Y, et al. Analysis of FOXO3 mutation in 114 Chinese women with premature ovarian failure. Reprod Biomed Online. 2010;20(4):499–503.
Marinakis NM, Tsoutsou E, Sofocleous C, Veltra D, Papaefthimiou P, Lytras A, et al. Ovarian insufficiency and secondary amenorrhea in a patient with a novel variant within GDF9 gene. Menopause. 2022;29(4):491–5.
Tucker EJ, Bell KM, Robevska G, van den Bergen J, Ayers KL, Listyasari N, et al. Meiotic genes in premature ovarian insufficiency: variants in HROB and REC8 as likely genetic causes. Eur J Hum Genet. 2022;30(2):219–28.
Wang B, Li L, Zhu Y, Zhang W, Wang X, Chen B, et al. Sequence variants of KHDRBS1 as high penetrance susceptibility risks for primary ovarian insufficiency by mis-regulating mRNA alternative splicing. Hum Reprod. 2017;32(10):2138–46.
Carlosama C, Elzaiat M, Patino LC, Mateus HE, Veitia RA, Laissue P. A homozygous donor splice-site mutation in the meiotic gene MSH4 causes primary ovarian insufficiency. Hum Mol Genet. 2017;26(16):3161–6.
Fievet A, Bellanger D, Zahed L, Burglen L, Derrien AC, Dubois d’Enghien C, et al. DNA repair functional analyses of NBN hypomorphic variants associated with NBN-related infertility. Hum Mutat. 2020;41(3):608–18.
Franca MM, Han X, Funari MFA, Lerario AM, Nishi MY, Fontenele EGP, et al. Exome Sequencing Reveals the POLR3H Gene as a Novel Cause of Primary Ovarian Insufficiency. J Clin Endocrinol Metab. 2019;104(7):2827–41.
Wang JL, Li SL, Qin YY, Chen ZJ. Analysis of progesterone receptor membrane component 1 mutation in Han Chinese women with premature ovarian failure. Reprod Biomed Online. 2014;29(5):640–3.
Sirchia F, Giorgio E, Cucinella L, Valente EM, Nappi RE. Biallelic mutations in PSMC3IP are associated with secondary amenorrhea: expanding the spectrum of premature ovarian insufficiency. J Assist Reprod Genet. 2022;39(5):1177–81.
Wang Q, Li D, Cai B, Chen Q, Li C, Wu Y, et al. Whole-exome sequencing reveals SALL4 variants in premature ovarian insufficiency: an update on genotype-phenotype correlations. Hum Genet. 2019;138(1):83–92.
Zhao S, Li G, Dalgleish R, Vujovic S, Jiao X, Li J, et al. Transcription factor SOHLH1 potentially associated with primary ovarian insufficiency. Fertil Steril. 2015;103(2):548–53 e5.
Hou D, Yao C, Xu B, Luo W, Ke H, Li Z, et al. Variations of C14ORF39 and SYCE1 Identified in Idiopathic Premature Ovarian Insufficiency and Nonobstructive Azoospermia. J Clin Endocrinol Metab. 2022;107(3):724–34.
Ramos L. WT1, NR0B1, NR5A1, LHX9, ZFP92, ZNF275, INSL3, and NRIP1 Genetic Variants in Patients with Premature Ovarian Insufficiency in a Mexican Cohort. Genes (Basel). 2022;13(4).
Ling L, Hou J, Liu D, Tang D, Zhang Y, Zeng Q, et al. Important role of the SDF-1/CXCR4 axis in the homing of systemically transplanted human amnion-derived mesenchymal stem cells (hAD-MSCs) to ovaries in rats with chemotherapy-induced premature ovarian insufficiency (POI). Stem Cell Res Ther. 2022;13(1):79.
Spears N, Lopes F, Stefansdottir A, Rossi V, De Felici M, Anderson RA, et al. Ovarian damage from chemotherapy and current approaches to its protection. Hum Reprod Update. 2019;25(6):673–93.
Qin X, Zhao Y, Zhang T, Yin C, Qiao J, Guo W, et al. TrkB agonist antibody ameliorates fertility deficits in aged and cyclophosphamide-induced premature ovarian failure model mice. Nat Commun. 2022;13(1):914.
Tammiste T, Kask K, Padrik P, Idla K, Rosenstein K, Jatsenko T, et al. A case report and follow-up of the first live birth after heterotopic transplantation of cryopreserved ovarian tissue in Eastern Europe. BMC Womens Health. 2019;19(1):65.
El-Derany MO, Said RS, El-Demerdash E. Bone Marrow-Derived Mesenchymal Stem Cells Reverse Radiotherapy-Induced Premature Ovarian Failure: Emphasis on Signal Integration of TGF-beta, Wnt/beta-Catenin and Hippo Pathways. Stem Cell Rev Rep. 2021;17(4):1429–45.
Mantawy EM, Said RS, Abdel-Aziz AK. Mechanistic approach of the inhibitory effect of chrysin on inflammatory and apoptotic events implicated in radiation-induced premature ovarian failure: Emphasis on TGF-beta/MAPKs signaling pathway. Biomed Pharmacother. 2019;109:293–303.
Dolmans MM, Hossay C, Nguyen TYT, Poirot C. Fertility Preservation: How to Preserve Ovarian Function in Children, Adolescents and Adults. J Clin Med. 2021;10(22).
Park EY, Hwang KH, Kim JH, Lee SH, Park KS, Choi SJ, et al. Epinephrine minimizes the use of bipolar coagulation and preserves ovarian reserve in laparoscopic ovarian cystectomy: a randomized controlled trial. Sci Rep. 2020;10(1):20911.
Park SJ, Seol A, Lee N, Lee S, Kim HS, Group PS 2021 A randomized controlled trial of ovarian reserve preservation and hemostasis during ovarian cystectomy Sci Rep 11 1 8495
Li F, Lu H, Huang Y, Wang X, Zhang Q, Li X, et al. A systematic review and meta-analysis of the association between Hashimoto’s thyroiditis and ovarian reserve. Int Immunopharmacol. 2022;108: 108670.
Vogt EC, Breivik L, Royrvik EC, Grytaas M, Husebye ES, Oksnes M. Primary Ovarian Insufficiency in Women With Addison’s Disease. J Clin Endocrinol Metab. 2021;106(7):e2656–63.
Khizroeva J, Nalli C, Bitsadze V, Lojacono A, Zatti S, Andreoli L, et al. Infertility in women with systemic autoimmune diseases. Best Pract Res Clin Endocrinol Metab. 2019;33(6): 101369.
Grossmann B, Saur S, Rall K, Pecher AC, Hubner S, Henes J, et al. Prevalence of autoimmune disease in women with premature ovarian failure. Eur J Contracept Reprod Health Care. 2020;25(1):72–5.
Szeliga A, Calik-Ksepka A, Maciejewska-Jeske M, Grymowicz M, Smolarczyk K, Kostrzak A, et al. Autoimmune Diseases in Patients with Premature Ovarian Insufficiency-Our Current State of Knowledge. Int J Mol Sci. 2021;22(5).
Fu X, Zheng Q, Zhang N, Ding M, Pan X, Wang W, et al. CUMS Promotes the Development of Premature Ovarian Insufficiency Mediated by Nerve Growth Factor and Its Receptor in Rats. Biomed Res Int. 2020;2020:1946853.
Rostami Dovom M, Noroozzadeh M, Mosaffa N, Zadeh-Vakili A, Piryaei A, Ramezani TF. Induced premature ovarian insufficiency by using D galactose and its effects on reproductive profiles in small laboratory animals: a systematic review. J Ovarian Res. 2019;12(1):96.
Lak R, Yazdizadeh B, Davari M, Nouhi M, Kelishadi R. Newborn screening for galactosaemia. Cochrane Database Syst Rev. 2020;6(6):CD012272.
Jin M, Yu Y, Huang H. An update on primary ovarian insufficiency. Sci China Life Sci. 2012;55(8):677–86.
Yang Y, Huang W, Yuan L. Effects of Environment and Lifestyle Factors on Premature Ovarian Failure. Adv Exp Med Biol. 2021;1300:63–111.
Patricia Wodi A, Marquez P, Mba-Jonas A, Barash F, Nguon K, Moro PL. Spontaneous reports of primary ovarian insufficiency after vaccination: A review of the vaccine adverse event reporting system (VAERS). Vaccine. 2023;41(9):1616–22.
Gong L, Ji HH, Tang XW, Pan LY, Chen X, Jia YT. Human papillomavirus vaccine-associated premature ovarian insufficiency and related adverse events: data mining of Vaccine Adverse Event Reporting System. Sci Rep. 2020;10(1):10762.
Tatang C, Arredondo Bisono T, Bergamasco A, Salvo F, Costa Clemens SA, Moride Y. Human Papillomavirus Vaccination and Premature Ovarian Failure: A Disproportionality Analysis Using the Vaccine Adverse Event Reporting System. Drugs Real World Outcomes. 2022;9(1):79–90.
Evangelinakis N, Geladari EV, Geladari CV, Kontogeorgi A, Papaioannou GK, Peppa M, et al. The influence of environmental factors on premature ovarian insufficiency and ovarian aging. Maturitas. 2024;179: 107871.
Gaskins AJ, Minguez-Alarcon L, Fong KC, Abdelmessih S, Coull BA, Chavarro JE, et al. Exposure to Fine Particulate Matter and Ovarian Reserve Among Women from a Fertility Clinic. Epidemiology. 2019;30(4):486–91.
Luderer U, Lim J, Ortiz L, Nguyen JD, Shin JH, Allen BD, et al. Exposure to environmentally relevant concentrations of ambient fine particulate matter (PM(2.5)) depletes the ovarian follicle reserve and causes sex-dependent cardiovascular changes in apolipoprotein E null mice. Part Fibre Toxicol. 2022;19(1):5.
Yang M, Tian F, Tao S, Xia M, Wang Y, Hu J, et al. Concentrated ambient fine particles exposure affects ovarian follicle development in mice. Ecotoxicol Environ Saf. 2022;231: 113178.
Liao BQ, Liu CB, Xie SJ, Liu Y, Deng YB, He SW, et al. Effects of fine particulate matter (PM(2.5)) on ovarian function and embryo quality in mice. Environ Int. 2020;135:105338.
Ding T, Yan W, Zhou T, Shen W, Wang T, Li M, et al. Endocrine disrupting chemicals impact on ovarian aging: Evidence from epidemiological and experimental evidence. Environ Pollut. 2022;305: 119269.
Kuai Y, Gao X, Yang H, Luo H, Xu Y, Liu C, et al. Pentachloronitrobenzene alters progesterone production and primordial follicle recruitment in cultured granulosa cells and rat ovarydagger. Biol Reprod. 2020;102(2):511–20.
Kaboli Kafshgiri S, Farkhondeh T, Miri-Moghaddam E. Glyphosate effects on the female reproductive systems: a systematic review. Rev Environ Health. 2022;37(4):487–500.
Jurewicz J, Radwan P, Wielgomas B, Radwan M, Karwacka A, Kaluzny P, et al. Exposure to pyrethroid pesticides and ovarian reserve. Environ Int. 2020;144:106028.
Jalouli M, Mofti A, Elnakady YA, Nahdi S, Feriani A, Alrezaki A, et al. Allethrin Promotes Apoptosis and Autophagy Associated with the Oxidative Stress-Related PI3K/AKT/mTOR Signaling Pathway in Developing Rat Ovaries. Int J Mol Sci. 2022;23(12).
Terayama H, Sakabe K, Kiyoshima D, Qu N, Sato T, Suyama K, et al. Effect of Neonicotinoid Pesticides on Japanese Water Systems: Review with Focus on Reproductive Toxicity. Int J Mol Sci. 2022;23(19).
He QK, Xu CL, Li YP, Xu ZR, Luo YS, Zhao SC, et al. Captan exposure disrupts ovarian homeostasis and affects oocytes quality via mitochondrial dysfunction induced apoptosis. Chemosphere. 2022;286(Pt 1):131625.
Li R, Xin B, Wang Q, Wang Z, Fu H, Yan Z, et al. Combined effect of unfolded protein response and circZc3h4, circRNA Scar in mouse ovary and uterus damage induced by procymidone. Ecotoxicol Environ Saf. 2022;229:113068.
Yun Y, Lee S, So C, Manhas R, Kim C, Wibowo T, et al. Oocyte Development and Quality in Young and Old Mice following Exposure to Atrazine. Environ Health Perspect. 2022;130(11):117007.
Schmidt S. Aged before Their Time: Atrazine and Diminished Egg Quality in Mice. Environ Health Perspect. 2022;130(12):124001.
An R, Wang X, Yang L, Zhang J, Wang N, Xu F, et al. Polystyrene microplastics cause granulosa cells apoptosis and fibrosis in ovary through oxidative stress in rats. Toxicology. 2021;449:152665.
Hou J, Lei Z, Cui L, Hou Y, Yang L, An R, et al. Polystyrene microplastics lead to pyroptosis and apoptosis of ovarian granulosa cells via NLRP3/Caspase-1 signaling pathway in rats. Ecotoxicol Environ Saf. 2021;212:112012.
Wu H, Liu Q, Yang N, Xu S. Polystyrene-microplastics and DEHP co-exposure induced DNA damage, cell cycle arrest and necroptosis of ovarian granulosa cells in mice by promoting ROS production. Sci Total Environ. 2023;871:161962.
Shen L, Liang C, Li D, Zhang Z, Wang X, Jiang T, et al. The association between exposure to multiple toxic metals and the risk of endometriosis: Evidence from the results of blood and follicular fluid. Sci Total Environ. 2023;855:158882.
Dutta S, Gorain B, Choudhury H, Roychoudhury S, Sengupta P. Environmental and occupational exposure of metals and female reproductive health. Environ Sci Pollut Res Int. 2022;29(41):62067–92.
Wang X, Ding N, Harlow SD, Randolph JF Jr, Mukherjee B, Gold EB, et al. Urinary metals and metal mixtures and timing of natural menopause in midlife women: The Study of Women’s Health Across the Nation. Environ Int. 2021;157:106781.
Pan W, Ye X, Zhu Z, Li C, Zhou J, Liu J. Urinary cadmium concentrations and risk of primary ovarian insufficiency in women: a case-control study. Environ Geochem Health. 2021;43(5):2025–35.
Budani MC, Tiboni GM. Ovotoxicity of cigarette smoke: A systematic review of the literature. Reprod Toxicol. 2017;72:164–81.
Chen H, Yang Y, Wang C, Hua X, Li H, Xie D, et al. Reproductive toxicity of UV-photodegraded polystyrene microplastics induced by DNA damage-dependent cell apoptosis in Caenorhabditis elegans. Sci Total Environ. 2022;811:152350.
Hua X, Feng X, Hua Y, Wang D. Paeoniflorin attenuates polystyrene nanoparticle-induced reduction in reproductive capacity and increase in germline apoptosis through suppressing DNA damage checkpoints in Caenorhabditis elegans. Sci Total Environ. 2023;871:162189.
Zhang Y, Wang X, Zhao Y, Zhao J, Yu T, Yao Y, et al. Reproductive toxicity of microplastics in female mice and their offspring from induction of oxidative stress. Environ Pollut. 2023;327: 121482.
Chatterjee A, Maity S, Banerjee S, Dutta S, Adhikari M, Guchhait R, et al. Toxicological impacts of nanopolystyrene on zebrafish oocyte with insight into the mechanism of action: An expression-based analysis. Sci Total Environ. 2022;830:154796.
Saeed A, Akhtar MF, Saleem A, Akhtar B, Sharif A. Reproductive and metabolic toxic effects of polystyrene microplastics in adult female Wistar rats: a mechanistic study. Environ Sci Pollut Res Int. 2023;30(22):63185–99.
Luo YS, He QK, Sun MX, Qiao FX, Liu YC, Xu CL, et al. Acrylonitrile exposure triggers ovarian inflammation and decreases oocyte quality probably via mitochondrial dysfunction induced apoptosis in mice. Chem Biol Interact. 2022;360:109934.
Liu Y, He QK, Xu ZR, Xu CL, Zhao SC, Luo YS, et al. Thiamethoxam Exposure Induces Endoplasmic Reticulum Stress and Affects Ovarian Function and Oocyte Development in Mice. J Agric Food Chem. 2021;69(6):1942–52.
Gastelbondo-Pastrana BI, Fernandes FH, Salvadori DMF, Santos DCD. The comet assay in Ceraeochrysa claveri (Neuroptera: Chrysopidae): A suitable approach for detecting somatic and germ cell genotoxicity induced by agrochemicals. Chemosphere. 2019;235:70–5.
Bhardwaj JK, Saraf P. Granulosa cell apoptosis by impairing antioxidant defense system and cellular integrity in caprine antral follicles post malathion exposure. Environ Toxicol. 2016;31(12):1944–54.
Wang W, Yang LL, Luo SM, Ma JY, Zhao Y, Shen W, et al. Toxic effects and possible mechanisms following malathion exposure in porcine granulosa cells. Environ Toxicol Pharmacol. 2018;64:172–80.
Bhardwaj JK, Mittal M, Saraf P, Sharma S. Ameliorative potential of vitamin C and E against Roundup-glyphosate induced genotoxicity triggering apoptosis in caprine granulosa cells. Environ Mol Mutagen. 2022;63(5):246–54.
Chitolina R, Nicola P, Sachett A, Bevilaqua F, Cunico L, Reginatto A, et al. Subacute exposure to Roundup(R) changes steroidogenesis and gene expression of the glutathione-glutaredoxin system in rat ovaries: Implications for ovarian toxicity of this glyphosate-based herbicide. Toxicol Appl Pharmacol. 2023;473: 116599.
Hamdaoui L, Naifar M, Rahmouni F, Harrabi B, Ayadi F, Sahnoun Z, et al. Subchronic exposure to kalach 360 SL-induced endocrine disruption and ovary damage in female rats. Arch Physiol Biochem. 2018;124(1):27–34.
Bhardwaj JK, Saraf P. N-acetyl-l-cysteine mediated regulation of DNA fragmentation, an apoptotic event, against methoxychlor toxicity in the granulosa cells of ovarian antral follicles. Mutat Res Genet Toxicol Environ Mutagen. 2020;858–860:503222.
Witek P, Grzesiak M, Koziorowski M, Slomczynska M, Knapczyk-Stwora K. Long-Term Changes in Ovarian Follicles of Gilts Exposed Neonatally to Methoxychlor: Effects on Oocyte-Derived Factors, Anti-Mullerian Hormone, Follicle-Stimulating Hormone, and Cognate Receptors. Int J Mol Sci. 2022;23(5).
Shah HK, Sharma T, Banerjee BD. Organochlorine pesticides induce inflammation, ROS production, and DNA damage in human epithelial ovary cells: An in vitro study. Chemosphere. 2020;246: 125691.
Wang W, Luo SM, Ma JY, Shen W, Yin S. Cytotoxicity and DNA Damage Caused from Diazinon Exposure by Inhibiting the PI3K-AKT Pathway in Porcine Ovarian Granulosa Cells. J Agric Food Chem. 2019;67(1):19–31.
Zhang Y, Xue W, Long R, Yang H, Wei W. Acetochlor affects zebrafish ovarian development by producing estrogen effects and inducing oxidative stress. Environ Sci Pollut Res Int. 2020;27(22):27688–96.
de Oliveira VS, Marins K, Mendes AKB, Zamoner A, Leite GAA, Silva F. In vivo exposure to pyriproxyfen causes ovarian oxidative stress and inhibits follicle maturation in zebrafish. J Appl Toxicol. 2023;43(6):799–807.
Bao J, Zhang Y, Wen R, Zhang L, Wang X. Low level of mancozeb exposure affects ovary in mice. Ecotoxicol Environ Saf. 2022;239: 113670.
Mzid M, Ghlissi Z, Salem MB, Khedir SB, Chaabouni K, Ayedi F, et al. Chemoprotective role of ethanol extract of Urtica urens L. against the toxicity of imidacloprid on endocrine disruption and ovarian morphometric in female rats, GC/MS analysis. Biomed Pharmacother. 2018;97:518–27.
Legoff L, Dali O, D’Cruz SC, Suglia A, Gely-Pernot A, Hemery C, et al. Ovarian dysfunction following prenatal exposure to an insecticide, chlordecone, associates with altered epigenetic features. Epigenetics Chromatin. 2019;12(1):29.
Hao Y, Zhang H, Zhang P, Yu S, Ma D, Li L, et al. Chlorothalonil inhibits mouse ovarian development through endocrine disruption. Toxicol Lett. 2019;303:38–47.
Kheirallah DAM, El-Samad LM, Abdel-Moneim AM. DNA damage and ovarian ultrastructural lesions induced by nickel oxide nano-particles in Blaps polycresta (Coleoptera: Tenebrionidae). Sci Total Environ. 2021;753:141743.
El-Samad LM, Hassan MA, Bakr NR, El-Ashram S, Radwan EH, Abdul Aziz KK, et al. Insights into Ag-NPs-mediated pathophysiology and ultrastructural aberrations in ovarian tissues of darkling beetles. Sci Rep. 2022;12(1):13899.
Tabandeh MR, Samie KA, Mobarakeh ES, Khadem MD, Jozaie S. Silver nanoparticles induce oxidative stress, apoptosis and impaired steroidogenesis in ovarian granulosa cells of cattle. Anim Reprod Sci. 2022;236: 106908.
Mojica-Vazquez LH, Madrigal-Zarraga D, Garcia-Martinez R, Boube M, Calderon-Segura ME, Oyallon J. Mercury chloride exposure induces DNA damage, reduces fertility, and alters somatic and germline cells in Drosophila melanogaster ovaries. Environ Sci Pollut Res Int. 2019;26(31):32322–32.
Ma Y, Zhu M, Miao L, Zhang X, Dong X, Zou X. Mercuric Chloride Induced Ovarian Oxidative Stress by Suppressing Nrf2-Keap1 Signal Pathway and its Downstream Genes in Laying Hens. Biol Trace Elem Res. 2018;185(1):185–96.
Ternjej I, Gaurina Srcek V, Mihaljevic Z, Kopjar N. Cytotoxic and genotoxic effects of water and sediment samples from gypsum mining area in channel catfish ovary (CCO) cells. Ecotoxicol Environ Saf. 2013;98:119–27.
Mansour FR, Nabiuni M, Amini E. Ovarian Toxicity Induced by Aluminum Chloride: Alteration of Cyp19a1, Pcna, Puma, and Map1lc3b genes Expression. Toxicology. 2022;466: 153084.
Qu J, Wang Q, Sun X, Li Y. The environment and female reproduction: Potential mechanism of cadmium poisoning to the growth and development of ovarian follicle. Ecotoxicol Environ Saf. 2022;244:114029.
Zhu M, Miao S, Zhou W, Elnesr SS, Dong X, Zou X. MAPK, AKT/FoxO3a and mTOR pathways are involved in cadmium regulating the cell cycle, proliferation and apoptosis of chicken follicular granulosa cells. Ecotoxicol Environ Saf. 2021;214: 112091.
Kechiche S, Venditti M, Knani L, Jablonska K, Dziegiel P, Messaoudi I, et al. First evidence of the protective role of melatonin in counteracting cadmium toxicity in the rat ovary via the mTOR pathway. Environ Pollut. 2021;270: 116056.
Xu G, Liu S, Huang M, Jiang X, Yang M. Cadmium induces apoptosis of human granulosa cell line KGN via mitochondrial dysfunction-mediated pathways. Ecotoxicol Environ Saf. 2021;220: 112341.
Zhang W, Wu T, Zhang C, Luo L, Xie M, Huang H. Cadmium exposure in newborn rats ovary induces developmental disorders of primordial follicles and the differential expression of SCF/c-kit gene. Toxicol Lett. 2017;280:20–8.
Sun Y, Liu Z, Zhang W, Lin H, Li Q, Liu C, et al. Paternal genetic effects of cadmium exposure during pregnancy on hormone synthesis disorders in ovarian granulosa cells of offspring. J Ovarian Res. 2023;16(1):98.
Ding H, Li Z, Li X, Yang X, Zhao J, Guo J, et al. FTO Alleviates CdCl(2)-Induced Apoptosis and Oxidative Stress via the AKT/Nrf2 Pathway in Bovine Granulosa Cells. Int J Mol Sci. 2022;23(9).
Luo L, Li J, Sun Y, Lv Y, Liu J, Li Y, et al. Maternal genetic intergenerational and transgenerational effects on hormone synthesis in ovarian granulosa cells of offspring exposed to cadmium during pregnancy. Ecotoxicol Environ Saf. 2023;263: 115278.
Mondal R, Pal P, Biswas S, Chattopadhyay A, Bandyopadhyay A, Mukhopadhyay A, et al. Attenuation of sodium arsenite mediated ovarian DNA damage, follicular atresia, and oxidative injury by combined application of vitamin E and C in post pubertal Wistar rats. Naunyn Schmiedebergs Arch Pharmacol. 2023;396(10):2701–20.
Nava-Rivera LE, Betancourt-Martinez ND, Lozoya-Martinez R, Carranza-Rosales P, Guzman-Delgado NE, Carranza-Torres IE, et al. Transgenerational effects in DNA methylation, genotoxicity and reproductive phenotype by chronic arsenic exposure. Sci Rep. 2021;11(1):8276.
Chen Y, Sun Y, Zhao A, Cai X, Yu A, Xu Q, et al. Arsenic exposure diminishes ovarian follicular reserve and induces abnormal steroidogenesis by DNA methylation. Ecotoxicol Environ Saf. 2022;241: 113816.
Anchordoquy JM, Anchordoquy JP, Nikoloff N, Pascua AM, Furnus CC. High copper concentrations produce genotoxicity and cytotoxicity in bovine cumulus cells. Environ Sci Pollut Res Int. 2017;24(24):20041–9.
Zou L, Cheng G, Xu C, Liu H, Wang Y, Li N, et al. Copper Nanoparticles Induce Oxidative Stress via the Heme Oxygenase 1 Signaling Pathway in vitro Studies. Int J Nanomedicine. 2021;16:1565–73.
Yiqin C, Yan S, Peiwen W, Yiwei G, Qi W, Qian X, et al. Copper exposure disrupts ovarian steroidogenesis in human ovarian granulosa cells via the FSHR/CYP19A1 pathway and alters methylation patterns on the SF-1 gene promoter. Toxicol Lett. 2022;356:11–20.
Wu Y, Yang R, Lan J, Wu Y, Huang J, Fan Q, et al. Iron overload modulates follicular microenvironment via ROS/HIF-1alpha/FSHR signaling. Free Radic Biol Med. 2023;196:37–52.
Feng Y, Yuan H, Wang W, Xu Y, Zhang J, Xu H, et al. Co-exposure to polystyrene microplastics and lead aggravated ovarian toxicity in female mice via the PERK/eIF2alpha signaling pathway. Ecotoxicol Environ Saf. 2022;243: 113966.
Trujillo-Vazquez SK, Gaona-Dominguez S, Villeda-Gonzalez JD, Librado-Osorio R, Luna-Lopez A, Bonilla-Gonzalez E, et al. Oxidative stress in the ovaries of mice chronically exposed to a low lead concentration: A generational approach. Reprod Toxicol. 2023;115:40–8.
Qu J, Niu H, Wang J, Wang Q, Li Y. Potential mechanism of lead poisoning to the growth and development of ovarian follicle. Toxicology. 2021;457: 152810.
Li F, Wang Y, Xu M, Hu N, Miao J, Zhao Y, et al. Single-nucleus RNA Sequencing reveals the mechanism of cigarette smoke exposure on diminished ovarian reserve in mice. Ecotoxicol Environ Saf. 2022;245:114093.
Konstantinidou F, Budani MC, Sarra A, Stuppia L, Tiboni GM, Gatta V. Impact of Cigarette Smoking on the Expression of Oxidative Stress-Related Genes in Cumulus Cells Retrieved from Healthy Women Undergoing IVF. Int J Mol Sci. 2021;22(23).
Budani MC, D’Aurora M, Stuppia L, Gatta V, Tiboni GM. Whole-body exposure to cigarette smoke alters oocyte miRNAs expression in C57BL/6 mice. Mol Reprod Dev. 2019;86(11):1741–57.
Chen T, Wu M, Dong Y, Kong B, Cai Y, Hei C, et al. Effect of e-cigarette refill liquid on follicular development and estrogen secretion in rats. Tob Induc Dis. 2022;20:36.
Budani MC, Gallorini M, Elsallabi O, Pino V, La Fratta I, Pesce M, et al. Cigarette smoke is associated with up-regulation of inducible NOS and COX-2 protein expression and activity in granulosa cells of women undergoing in vitro fertilization. Reprod Toxicol. 2022;113:128–35.
Chen KM, Sun YW, Cooper TK, Benitez G, Aliaga C, Zhu J, et al. Comparative Tumorigenicity and DNA Damage Induced by Dibenzo[def,p]chrysene and Its Metabolites in the Mouse Ovary. Chem Res Toxicol. 2018;31(11):1111–8.
Cheng SF, Qin XS, Han ZL, Sun XF, Feng YN, Yang F, et al. Nicotine exposure impairs germ cell development in human fetal ovaries cultured in vitro. Aging (Albany NY). 2018;10(7):1556–74.
Wang YF, Sun XF, Han ZL, Li L, Ge W, Zhao Y, et al. Protective effects of melatonin against nicotine-induced disorder of mouse early folliculogenesis. Aging (Albany NY). 2018;10(3):463–80.
Fan G, Zhang Q, Wan Y, Lv F, Chen Y, Ni Y, et al. Decreased levels of H3K9ac and H3K27ac in the promotor region of ovarian P450 aromatase mediated low estradiol synthesis in female offspring rats induced by prenatal nicotine exposure as well as in human granulosa cells after nicotine treatment. Food Chem Toxicol. 2019;128:256–66.
Sezer Z, Ekiz Yilmaz T, Gungor ZB, Kalay F, Guzel E. Effects of vitamin E on nicotine-induced lipid peroxidation in rat granulosa cells: Folliculogenesis. Reprod Biol. 2020;20(1):63–74.
Faghani M, Saedi S, Khanaki K, Mohammadghasemi F. Ginseng alleviates folliculogenesis disorders via induction of cell proliferation and downregulation of apoptotic markers in nicotine-treated mice. J Ovarian Res. 2022;15(1):14.
He QK, Li YP, Xu ZR, Wei WB, Qiao FX, Sun MX, et al. 3-MCPD exposure enhances ovarian fibrosis and reduces oocyte quality in mice. Environ Pollut. 2023;316(Pt 2): 120662.
Onder GO, Goktepe O, Karaman E, Karakas E, Mat OC, Bolat D, et al. Nonylphenol Exposure-Induced Oocyte Quality Deterioration Could be Reversed by Boric Acid Supplementation in Rats. Biol Trace Elem Res. 2023;201(9):4518–29.
Song W, Qiu YT, Li XZ, Sun QY, Chen LN. 4-vinylcyclohexene diepoxide induces apoptosis by excessive reactive oxygen species and DNA damage in human ovarian granulosa cells. Toxicol In Vitro. 2023;91:105613.
Li Y, Li M, Liu J, Nie G, Yang H. Altered m6A modification is involved YAP-mediated apoptosis response in 4-vinylcyclohexene diepoxide induced ovotoxicity. Ecotoxicol Environ Saf. 2023;262: 115192.
Zhou Q, Jin X, Wang J, Li H, Yang L, Wu W, et al. 4-vinylcyclohexene diepoxide induces premature ovarian insufficiency in rats by triggering the autophagy of granule cells through regulating miR-144. J Reprod Immunol. 2023;157: 103928.
Liu X, Craig ZR. Environmentally relevant exposure to dibutyl phthalate disrupts DNA damage repair gene expression in the mouse ovarydagger. Biol Reprod. 2019;101(4):854–67.
Tu Z, Mu X, Chen X, Geng Y, Zhang Y, Li Q, et al. Dibutyl phthalate exposure disrupts the progression of meiotic prophase I by interfering with homologous recombination in fetal mouse oocytes. Environ Pollut. 2019;252(Pt A):388–98.
Ganesan S, Keating AF. Bisphenol A-Induced Ovotoxicity Involves DNA Damage Induction to Which the Ovary Mounts a Protective Response Indicated by Increased Expression of Proteins Involved in DNA Repair and Xenobiotic Biotransformation. Toxicol Sci. 2016;152(1):169–80.
Zhang M, Dai X, Lu Y, Miao Y, Zhou C, Cui Z, et al. Melatonin protects oocyte quality from Bisphenol A-induced deterioration in the mouse. J Pineal Res. 2017;62(3).
Huang M, Liu S, Fu L, Jiang X, Yang M. Bisphenol A and its analogues bisphenol S, bisphenol F and bisphenol AF induce oxidative stress and biomacromolecular damage in human granulosa KGN cells. Chemosphere. 2020;253: 126707.
Huang M, Huang M, Li X, Liu S, Fu L, Jiang X, et al. Bisphenol A induces apoptosis through GPER-dependent activation of the ROS/Ca(2+)-ASK1-JNK pathway in human granulosa cell line KGN. Ecotoxicol Environ Saf. 2021;208: 111429.
Liu Y, Wang L, Zhu L, Ran B, Wang Z. Bisphenol A disturbs transcription of steroidogenic genes in ovary of rare minnow Gobiocypris rarus via the abnormal DNA and histone methylation. Chemosphere. 2020;240: 124935.
Zhu L, Liu Y, Xue X, Yuan C, Wang Z. BPA’s transgenerational disturbance to transcription of ovarian steroidogenic genes in rare minnow Gobiocypris rarus via DNA and histone methylation. Sci Total Environ. 2021;762: 143055.
Lopez-Rodriguez D, Franssen D, Bakker J, Lomniczi A, Parent AS. Cellular and molecular features of EDC exposure: consequences for the GnRH network. Nat Rev Endocrinol. 2021;17(2):83–96.
Meng Y, Yannan Z, Ren L, Qi S, Wei W, Lihong J. Adverse reproductive function induced by maternal BPA exposure is associated with abnormal autophagy and activating inflamation via mTOR and TLR4/NF-kappaB signaling pathways in female offspring rats. Reprod Toxicol. 2020;96:185–94.
Prokesova S, Ghaibour K, Liska F, Klein P, Fenclova T, Stiavnicka M, et al. Acute low-dose bisphenol S exposure affects mouse oocyte quality. Reprod Toxicol. 2020;93:19–27.
Pal S, Sahu A, Verma R, Haldar C. BPS-induced ovarian dysfunction: Protective actions of melatonin via modulation of SIRT-1/Nrf2/NFkB and IR/PI3K/pAkt/GLUT-4 expressions in adult golden hamster. J Pineal Res. 2023;75(1): e12869.
Jia Z, Wang H, Feng Z, Zhang S, Wang L, Zhang J, et al. Fluorene-9-bisphenol exposure induces cytotoxicity in mouse oocytes and causes ovarian damage. Ecotoxicol Environ Saf. 2019;180:168–78.
Jeong PS, Lee S, Park SH, Kim MJ, Kang HG, Nanjidsuren T, et al. Butylparaben Is Toxic to Porcine Oocyte Maturation and Subsequent Embryonic Development Following In Vitro Fertilization. Int J Mol Sci. 2020;21(10).
Estefania Gonzalez-Alvarez M, Severin A, Sayadi M, Keating AF. PFOA-Induced Ovotoxicity Differs Between Lean and Obese Mice With Impacts on Ovarian Reproductive and DNA Damage Sensing and Repair Proteins. Toxicol Sci. 2022;190(2):173–88.
Zhou YT, Li R, Li SH, Ma X, Liu L, Niu D, et al. Perfluorooctanoic acid (PFOA) exposure affects early embryonic development and offspring oocyte quality via inducing mitochondrial dysfunction. Environ Int. 2022;167:107413.
Basini G, Bussolati S, Torcianti V, Grasselli F. Perfluorooctanoic Acid (PFOA) Induces Redox Status Disruption in Swine Granulosa Cells. Vet Sci. 2022;9(6).
Lopez-Arellano P, Lopez-Arellano K, Luna J, Flores D, Jimenez-Salazar J, Gavia G, et al. Perfluorooctanoic acid disrupts gap junction intercellular communication and induces reactive oxygen species formation and apoptosis in mouse ovaries. Environ Toxicol. 2019;34(1):92–8.
Yang M, Lee Y, Gao L, Chiu K, Meling DD, Flaws JA, et al. Perfluorooctanoic Acid Disrupts Ovarian Steroidogenesis and Folliculogenesis in Adult Mice. Toxicol Sci. 2022;186(2):260–8.
Liu JC, Xing CH, Xu Y, Pan ZN, Zhang HL, Zhang Y, et al. DEHP exposure to lactating mice affects ovarian hormone production and antral follicle development of offspring. J Hazard Mater. 2021;416:125862.
Li MH, Wang JJ, Feng YQ, Liu X, Yan ZH, Zhang XJ, et al. H3K4me3 as a target of di(2-ethylhexyl) phthalate (DEHP) impairing primordial follicle assembly. Chemosphere. 2023;310: 136811.
Tripathi A, Pandey V, Sahu AN, Singh A, Dubey PK. Di-(2-ethylhexyl) phthalate (DEHP) inhibits steroidogenesis and induces mitochondria-ROS mediated apoptosis in rat ovarian granulosa cells. Toxicol Res (Camb). 2019;8(3):381–94.
Sun J, Gan L, Lv S, Wang T, Dai C, Sun J. Exposure to Di-(2-Ethylhexyl) phthalate drives ovarian dysfunction by inducing granulosa cell pyroptosis via the SLC39A5/NF-kappaB/NLRP3 axis. Ecotoxicol Environ Saf. 2023;252: 114625.
Fu X, Han H, Li Y, Xu B, Dai W, Zhang Y, et al. Di-(2-ethylhexyl) phthalate exposure induces female reproductive toxicity and alters the intestinal microbiota community structure and fecal metabolite profile in mice. Environ Toxicol. 2021;36(6):1226–42.
Yan W, Li M, Guo Q, Li X, Zhou S, Dai J, et al. Chronic exposure to propylparaben at the humanly relevant dose triggers ovarian aging in adult mice. Ecotoxicol Environ Saf. 2022;235: 113432.
Li M, Zhou S, Wu Y, Li Y, Yan W, Guo Q, et al. Prenatal exposure to propylparaben at human-relevant doses accelerates ovarian aging in adult mice. Environ Pollut. 2021;285: 117254.
Martinez-Quezada R, Gonzalez-Castaneda G, Bahena I, Dominguez A, Dominguez-Lopez P, Casas E, et al. Effect of perfluorohexane sulfonate on pig oocyte maturation, gap-junctional intercellular communication, mitochondrial membrane potential and DNA damage in cumulus cells in vitro. Toxicol In Vitro. 2021;70:105011.
Wang W, Zhao M, Zhao Y, Shen W, Yin S. PDGFRalpha/beta-PI3K-Akt pathway response to the interplay of mitochondrial dysfunction and DNA damage in Aroclor 1254-exposed porcine granulosa cells. Environ Pollut. 2020;263(Pt A):114534.
Matikainen T, Perez GI, Jurisicova A, Pru JK, Schlezinger JJ, Ryu HY, et al. Aromatic hydrocarbon receptor-driven Bax gene expression is required for premature ovarian failure caused by biohazardous environmental chemicals. Nat Genet. 2001;28(4):355–60.
Zhou S, Xi Y, Chen Y, Zhang Z, Wu C, Yan W, et al. Ovarian Dysfunction Induced by Chronic Whole-Body PM2.5 Exposure. Small. 2020;16(33):e2000845.
Prieto-Huecas L, Piera-Jordan CA, Serrano De La Cruz-Delgado V, Zaragoza-Marti A, Garcia-Velert MB, Tordera-Terrades C, et al. Assessment of Nutritional Status and Its Influence on Ovarian Reserve: A Systematic Review. Nutrients. 2023;15(10).
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):172–89.
Cavalcante MB, Sampaio OGM, Camara FEA, Schneider A, de Avila BM, Prosczek J, et al. Ovarian aging in humans: potential strategies for extending reproductive lifespan. Geroscience. 2023;45(4):2121–33.
Johnson J, Canning J, Kaneko T, Pru JK, Tilly JL. Germline stem cells and follicular renewal in the postnatal mammalian ovary. Nature. 2004;428(6979):145–50.
Baker TG. A Quantitative and Cytological Study of Germ Cells in Human Ovaries. Proc R Soc Lond B Biol Sci. 1963;158:417–33.
Adhikari D, Liu K. Molecular mechanisms underlying the activation of mammalian primordial follicles. Endocr Rev. 2009;30(5):438–64.
Ford EA, Beckett EL, Roman SD, McLaughlin EA, Sutherland JM. Advances in human primordial follicle activation and premature ovarian insufficiency. Reproduction. 2020;159(1):R15–29.
Davis CK, Vemuganti R. DNA damage and repair following traumatic brain injury. Neurobiol Dis. 2021;147:105143.
Takahashi A, Yousif A, Hong L, Chefetz I. Premature ovarian insufficiency: pathogenesis and therapeutic potential of mesenchymal stem cell. J Mol Med (Berl). 2021;99(5):637–50.
Ding X, Gong X, Fan Y, Cao J, Zhao J, Zhang Y, et al. DNA double-strand break genetic variants in patients with premature ovarian insufficiency. J Ovarian Res. 2023;16(1):135.
Day FR, Ruth KS, Thompson DJ, Lunetta KL, Pervjakova N, Chasman DI, et al. Large-scale genomic analyses link reproductive aging to hypothalamic signaling, breast cancer susceptibility and BRCA1-mediated DNA repair. Nat Genet. 2015;47(11):1294–303.
Turan V, Oktay K. BRCA-related ATM-mediated DNA double-strand break repair and ovarian aging. Hum Reprod Update. 2020;26(1):43–57.
Carusillo A, Mussolino C. DNA Damage: From Threat to Treatment. Cells. 2020;9(7).
Shadfar S, Parakh S, Jamali MS, Atkin JD. Redox dysregulation as a driver for DNA damage and its relationship to neurodegenerative diseases. Transl Neurodegener. 2023;12(1):18.
Garaycoechea JI, Crossan GP, Langevin F, Mulderrig L, Louzada S, Yang F, et al. Alcohol and endogenous aldehydes damage chromosomes and mutate stem cells. Nature. 2018;553(7687):171–7.
Petr MA, Tulika T, Carmona-Marin LM, Scheibye-Knudsen M. Protecting the Aging Genome. Trends Cell Biol. 2020;30(2):117–32.
Zheng X, Zhang W, Hu Y, Zhao Z, Wu J, Zhang X, et al. DNA repair byproduct 8-oxoguanine base promotes myoblast differentiation. Redox Biol. 2023;61:102634.
Li C, Zhu H, Jin S, Maksoud LM, Jain N, Sun J, et al. Structural basis of DNA polymerase theta mediated DNA end joining. Nucleic Acids Res. 2023;51(1):463–74.
Gonfloni S, Jodice C, Gustavino B, Valentini E. DNA Damage Stress Response and Follicle Activation: Signaling Routes of Mammalian Ovarian Reserve. Int J Mol Sci. 2022;23(22).
Puy V, Barroca V, Messiaen S, Menard V, Torres C, Devanand C, et al. Mouse model of radiation-induced premature ovarian insufficiency reveals compromised oocyte quality: implications for fertility preservation. Reprod Biomed Online. 2021;43(5):799–809.
Tamari T, Kawar-Jaraisy R, Doppelt O, Giladi B, Sabbah N, Zigdon-Giladi H. The Paracrine Role of Endothelial Cells in Bone Formation via CXCR4/SDF-1 Pathway. Cells. 2020;9(6):1325.
Li X, Zang N, Zhang N, Pang L, Lv L, Meng X, et al. DNA damage resulting from human endocrine disrupting chemical exposure: Genotoxicity, detection and dietary phytochemical intervention. Chemosphere. 2023;338:139522.
Nguyen QN, Zerafa N, Liew SH, Findlay JK, Hickey M, Hutt KJ. Cisplatin- and cyclophosphamide-induced primordial follicle depletion is caused by direct damage to oocytes. Mol Hum Reprod. 2019;25(8):433–44.
Tuppi M, Kehrloesser S, Coutandin DW, Rossi V, Luh LM, Strubel A, et al. Oocyte DNA damage quality control requires consecutive interplay of CHK2 and CK1 to activate p63. Nat Struct Mol Biol. 2018;25(3):261–9.
Francisco LFV, Baldivia DDS, Crispim BDA, Klafke S, Castilho PF, Viana LF, et al. Acute Toxic and Genotoxic Effects of Aluminum and Manganese Using In Vitro Models. Toxics. 2021;9(7).
Quezada-Maldonado EM, Sanchez-Perez Y, Chirino YI, Garcia-Cuellar CM. Airborne particulate matter induces oxidative damage, DNA adduct formation and alterations in DNA repair pathways. Environ Pollut. 2021;287:117313.
Liu JC, Lai FN, Li L, Sun XF, Cheng SF, Ge W, et al. Di (2-ethylhexyl) phthalate exposure impairs meiotic progression and DNA damage repair in fetal mouse oocytes in vitro. Cell Death Dis. 2017;8(8):e2966.
Zeng H, Li M, Hua Q, Liu Y, Shao Y, Diao Q, et al. Circular RNA circ_Cabin1 promotes DNA damage in multiple mouse organs via inhibition of non-homologous end-joining repair upon PM(2.5) exposure. Arch Toxicol. 2021;95(10):3235–51.
Clarke TL, Mostoslavsky R. DNA repair as a shared hallmark in cancer and ageing. Mol Oncol. 2022;16(18):3352–79.
Tadesse S, Kidane D, Guller S, Luo T, Norwitz NG, Arcuri F, et al. In vivo and in vitro evidence for placental DNA damage in preeclampsia. PLoS ONE. 2014;9(1):e86791.
Liu M, Zeng T, Zhang X, Liu C, Wu Z, Yao L, et al. ATR/Chk1 signaling induces autophagy through sumoylated RhoB-mediated lysosomal translocation of TSC2 after DNA damage. Nat Commun. 2018;9(1):4139.
Yang L, Xu LZ, Liu ZQ, Yang G, Geng XR, Mo LH, et al. Interleukin-13 interferes with activation-induced t-cell apoptosis by repressing p53 expression. Cell Mol Immunol. 2016;13(5):669–77.
Leon-Reyes G, Argoty-Pantoja AD, Becerra-Cervera A, Lopez-Montoya P, Rivera-Paredez B, Velazquez-Cruz R. Oxidative-Stress-Related Genes in Osteoporosis: A Systematic Review. Antioxidants (Basel). 2023;12(4).
Wang L, Tang J, Wang L, Tan F, Song H, Zhou J, et al. Oxidative stress in oocyte aging and female reproduction. J Cell Physiol. 2021;236(12):7966–83.
Shi X, Tian Y, Zhai S, Liu Y, Chu S, Xiong Z. The progress of research on the application of redox nanomaterials in disease therapy. Front Chem. 2023;11:1115440.
Tsui KH, Li CJ. Mitoquinone shifts energy metabolism to reduce ROS-induced oxeiptosis in female granulosa cells and mouse oocytes. Aging (Albany NY). 2023;15(1):246–60.
Khadrawy O, Gebremedhn S, Salilew-Wondim D, Taqi MO, Neuhoff C, Tholen E, et al. Endogenous and Exogenous Modulation of Nrf2 Mediated Oxidative Stress Response in Bovine Granulosa Cells: Potential Implication for Ovarian Function. Int J Mol Sci. 2019;20(7).
Shi YQ, Zhu XT, Zhang SN, Ma YF, Han YH, Jiang Y, et al. Premature ovarian insufficiency: a review on the role of oxidative stress and the application of antioxidants. Front Endocrinol (Lausanne). 2023;14:1172481.
Zhang X, Zhou C, Li W, Li J, Wu W, Tao J, et al. Vitamin C Protects Porcine Oocytes From Microcystin-LR Toxicity During Maturation. Front Cell Dev Biol. 2020;8:582715.
Bt Hj Idrus R, Sainik N, Nordin A, Saim AB, Sulaiman N. Cardioprotective Effects of Honey and Its Constituent: An Evidence-Based Review of Laboratory Studies and Clinical Trials. Int J Environ Res Public Health. 2020;17(10).
Chaudhary GR, Yadav PK, Yadav AK, Tiwari M, Gupta A, Sharma A, et al. Necroptosis in stressed ovary. J Biomed Sci. 2019;26(1):11.
Zhang S, Liu Q, Chang M, Pan Y, Yahaya BH, Liu Y, et al. Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis. Cell Death Dis. 2023;14(5):340.
Li BL, Liu X, Gao M, Zhang F, Chen X, He Z, et al. Programmed NP Cell Death Induced by Mitochondrial ROS in a One-Strike Loading Disc Degeneration Organ Culture Model. Oxid Med Cell Longev. 2021;2021:5608133.
Wang Y, Chen CZ, Fu XH, Liu JB, Peng YX, Wang YJ, et al. CPEB3 regulates the proliferation and apoptosis of bovine cumulus cells. Anim Sci J. 2020;91(1):e13416.
Wang X, Wang QC, Sun Z, Li T, Yang K, An C, et al. ER stress mediated degradation of diacylglycerol acyltransferase impairs mitochondrial functions in TMCO1 deficient cells. Biochem Biophys Res Commun. 2019;512(4):914–20.
Li W, Liu Z, Wang P, Di R, Wang X, Liu Y, et al. The transcription factor RUNX1 affects the maturation of porcine oocytes via the BMP15/TGF-beta signaling pathway. Int J Biol Macromol. 2023;238:124026.
He WB, Du J, Yang XW, Li W, Tang WL, Dai C, et al. Novel inactivating mutations in the FSH receptor cause premature ovarian insufficiency with resistant ovary syndrome. Reprod Biomed Online. 2019;38(3):397–406.
Liu J, Yang Y, He Y, Feng C, Ou H, Yang J, et al. Erxian decoction alleviates cisplatin-induced premature ovarian failure in rats by reducing oxidation levels in ovarian granulosa cells. J Ethnopharmacol. 2023;304:116046.
Huang B, Qian C, Ding C, Meng Q, Zou Q, Li H. Fetal liver mesenchymal stem cells restore ovarian function in premature ovarian insufficiency by targeting MT1. Stem Cell Res Ther. 2019;10(1):362.
Hao X, Anastacio A, Liu K, Rodriguez-Wallberg KA. Ovarian Follicle Depletion Induced by Chemotherapy and the Investigational Stages of Potential Fertility-Protective Treatments-A Review. Int J Mol Sci. 2019;20(19).
Mantawy EM, Said RS, Kassem DH, Abdel-Aziz AK, Badr AM. Novel molecular mechanisms underlying the ameliorative effect of N-acetyl-L-cysteine against Upsilon-radiation-induced premature ovarian failure in rats. Ecotoxicol Environ Saf. 2020;206:111190.
Haddad YH, Said RS, Kamel R, Morsy EME, El-Demerdash E. Phytoestrogen genistein hinders ovarian oxidative damage and apoptotic cell death-induced by ionizing radiation: co-operative role of ER-beta, TGF-beta, and FOXL-2. Sci Rep. 2020;10(1):13551.
He L, Long X, Yu N, Li Y, Liu X, Cheng X. Premature Ovarian Insufficiency (POI) Induced by Dynamic Intensity Modulated Radiation Therapy via P13K-AKT-FOXO3a in Rat Models. Biomed Res Int. 2021;2021:7273846.
de Oliveira AN, Martins Pereira G, Di Domenico M, Costanzo G, Benevenuto S, de Oliveira Fonoff AM, et al. Inflammation response, oxidative stress and DNA damage caused by urban air pollution exposure increase in the lack of DNA repair XPC protein. Environ Int. 2020;145: 106150.
Chen Y, Xi Y, Li M, Wu Y, Yan W, Dai J, et al. Maternal exposure to PM2.5 decreases ovarian reserve in neonatal offspring mice through activating PI3K/AKT/FoxO3a pathway and ROS-dependent NF-kappaB pathway. Toxicology. 2022;481:153352.
Das A. The emerging role of microplastics in systemic toxicity: Involvement of reactive oxygen species (ROS). Sci Total Environ. 2023;895: 165076.
da Costa CS, Oliveira TF, Freitas-Lima LC, Padilha AS, Krause M, Carneiro M, et al. Subacute cadmium exposure disrupts the hypothalamic-pituitary-gonadal axis, leading to polycystic ovarian syndrome and premature ovarian failure features in female rats. Environ Pollut. 2021;269: 116154.
Patel UN, Patel UD, Khadayata AV, Vaja RK, Modi CM, Patel HB. Long-term exposure of the binary mixture of cadmium and mercury damages the developed ovary of adult zebrafish. Environ Sci Pollut Res Int. 2022;29(29):44928–38.
Basini G, Bussolati S, Torcianti V, Grasselli F. Perfluorooctanoic acid (PFOA) affects steroidogenesis and antioxidant defence in granulosa cells from swine ovary. Environ Toxicol Pharmacol. 2023;101: 104169.
Shi L, Zhang J, Lai Z, Tian Y, Fang L, Wu M, et al. Long-Term Moderate Oxidative Stress Decreased Ovarian Reproductive Function by Reducing Follicle Quality and Progesterone Production. PLoS ONE. 2016;11(9): e0162194.
Huang E, Chen L. RNA N(6)-methyladenosine modification in female reproductive biology and pathophysiology. Cell Commun Signal. 2023;21(1):53.
Eslami H, Eslami A, Favaedi R, Asadpour U, Zari Moradi S, Eftekhari-Yazdi P, et al. Epigenetic Aberration of FMR1 Gene in Infertile Women with Diminished Ovarian Reserve. Cell J. 2018;20(1):78–83.
Roy S, Sinha N, Huang B, Cline-Fedewa H, Gleicher N, Wang J, et al. Jumonji Domain-containing Protein-3 (JMJD3/Kdm6b) Is Critical for Normal Ovarian Function and Female Fertility. Endocrinology. 2022;163(5).
Yang W, Ma Y, Jin J, Ren P, Zhou H, Xu S, et al. Cyclophosphamide Exposure Causes Long-Term Detrimental Effect of Oocytes Developmental Competence Through Affecting the Epigenetic Modification and Maternal Factors’ Transcription During Oocyte Growth. Front Cell Dev Biol. 2021;9: 682060.
Di Emidio G, D’Aurora M, Placidi M, Franchi S, Rossi G, Stuppia L, et al. Pre-conceptional maternal exposure to cyclophosphamide results in modifications of DNA methylation in F1 and F2 mouse oocytes: evidence for transgenerational effects. Epigenetics. 2019;14(11):1057–64.
Leung CT, Yang Y, Yu KN, Tam N, Chan TF, Lin X, et al. Low-Dose Radiation Can Cause Epigenetic Alterations Associated With Impairments in Both Male and Female Reproductive Cells. Front Genet. 2021;12: 710143.
Panay N, Anderson RA, Nappi RE, Vincent AJ, Vujovic S, Webber L, et al. Premature ovarian insufficiency: an International Menopause Society White Paper. Climacteric. 2020;23(5):426–46.
La Merrill MA, Vandenberg LN, Smith MT, Goodson W, Browne P, Patisaul HB, et al. Consensus on the key characteristics of endocrine-disrupting chemicals as a basis for hazard identification. Nat Rev Endocrinol. 2020;16(1):45–57.
Ge W, Li L, Dyce PW, De Felici M, Shen W. Establishment and depletion of the ovarian reserve: physiology and impact of environmental chemicals. Cell Mol Life Sci. 2019;76(9):1729–46.
Loganathan N, Salehi A, Chalmers JA, Belsham DD. Bisphenol A Alters Bmal1, Per2, and Rev-Erba mRNA and Requires Bmal1 to Increase Neuropeptide Y Expression in Hypothalamic Neurons. Endocrinology. 2019;160(1):181–92.
Li X, Li X, Deng L. Chrysin reduces inflammation and oxidative stress and improves ovarian function in D-gal-induced premature ovarian failure. Bioengineered. 2022;13(4):8291–301.
Ibrahim MA, Albahlol IA, Wani FA, Abd-Eltawab Tammam A, Kelleni MT, Sayeed MU, et al. Resveratrol protects against cisplatin-induced ovarian and uterine toxicity in female rats by attenuating oxidative stress, inflammation and apoptosis. Chem Biol Interact. 2021;338: 109402.
Jiao X, Zhang X, Li N, Zhang D, Zhao S, Dang Y, et al. T(reg) deficiency-mediated T(H) 1 response causes human premature ovarian insufficiency through apoptosis and steroidogenesis dysfunction of granulosa cells. Clin Transl Med. 2021;11(6): e448.
Ali I, Padhiar AA, Wang T, He L, Chen M, Wu S, et al. Stem Cell-Based Therapeutic Strategies for Premature Ovarian Insufficiency and Infertility: A Focus on Aging. Cells. 2022;11(23).
Huang Y, Hu C, Ye H, Luo R, Fu X, Li X, et al. Inflamm-Aging: A New Mechanism Affecting Premature Ovarian Insufficiency. J Immunol Res. 2019;2019:8069898.
Hahn J, Cook NR, Alexander EK, Friedman S, Walter J, Bubes V, et al. Vitamin D and marine omega 3 fatty acid supplementation and incident autoimmune disease: VITAL randomized controlled trial. BMJ. 2022;376: e066452.
Zielinski MR, Systrom DM, Rose NR. Fatigue, Sleep, and Autoimmune and Related Disorders. Front Immunol. 2019;10:1827.
Al-Shahat A, Hulail MAE, Soliman NMM, Khamis T, Fericean LM, Arisha AH, et al. Melatonin Mitigates Cisplatin-Induced Ovarian Dysfunction via Altering Steroidogenesis, Inflammation, Apoptosis, Oxidative Stress, and PTEN/PI3K/Akt/mTOR/AMPK Signaling Pathway in Female Rats. Pharmaceutics. 2022;14(12).
Guo RB, Wang GF, Zhao AP, Gu J, Sun XL, Hu G. Paeoniflorin protects against ischemia-induced brain damages in rats via inhibiting MAPKs/NF-kappaB-mediated inflammatory responses. PLoS ONE. 2012;7(11): e49701.
Zhang Q, Wei Z, Weng H, Chen Y, Zhang J, Mei S, et al. Folic Acid Preconditioning Alleviated Radiation-Induced Ovarian Dysfunction in Female Mice. Front Nutr. 2022;9: 854655.
Akdemir Y, Akpolat M, Elmas O, Kececi M, Buyukuysal C, Cetinkaya B, et al. Capsaicin prevents radiotherapy-induced premature ovarian failure in rats. Reprod Fertil Dev. 2022;34(3):350–61.
Barberino RS, Lins T, Monte APO, Silva RLS, Andrade KO, Campinho DSP, et al. Epigallocatechin-3-gallate attenuates cyclophosphamide-induced damage in mouse ovarian tissue via suppressing inflammation, apoptosis, and expression of phosphorylated Akt, FOXO3a and rpS6. Reprod Toxicol. 2022;113:42–51.
Stringer JM, Alesi LR, Winship AL, Hutt KJ. Beyond apoptosis: evidence of other regulated cell death pathways in the ovary throughout development and life. Hum Reprod Update. 2023;29(4):434–56.
Dey P, Luciano AM. A century of programmed cell death in the ovary: a commentary. J Assist Reprod Genet. 2022;39(1):63–6.
Raducka-Jaszul O, Boguslawska DM, Jedruchniewicz N, Sikorski AF. Role of Extrinsic Apoptotic Signaling Pathway during Definitive Erythropoiesis in Normal Patients and in Patients with beta-Thalassemia. Int J Mol Sci. 2020;21(9).
Hojo T, Skarzynski DJ, Okuda K. Apoptosis, autophagic cell death, and necroptosis: different types of programmed cell death in bovine corpus luteum regression. J Reprod Dev. 2022;68(6):355–60.
Zhang T, Ma Q, Shen Q, Jiang C, Zou F, Shen Y, et al. Identification of novel biallelic variants in BMP15 in two siblings with premature ovarian insufficiency. J Assist Reprod Genet. 2022;39(9):2125–34.
Xue W, Xue F, Jia T, Hao A. Research and experimental verification of the molecular mechanism of berberine in improving premature ovarian failure based on network pharmacology. Bioengineered. 2022;13(4):9885–900.
Du R, Cheng X, Ji J, Lu Y, Xie Y, Wang W, et al. Mechanism of ferroptosis in a rat model of premature ovarian insufficiency induced by cisplatin. Sci Rep. 2023;13(1):4463.
Xie QE, Wang MY, Cao ZP, Du X, Ji DM, Liang D, et al. Melatonin protects against excessive autophagy-induced mitochondrial and ovarian reserve function deficiency though ERK signaling pathway in Chinese hamster ovary (CHO) cells. Mitochondrion. 2021;61:44–53.
Chen Y, Zhao Y, Miao C, Yang L, Wang R, Chen B, et al. Quercetin alleviates cyclophosphamide-induced premature ovarian insufficiency in mice by reducing mitochondrial oxidative stress and pyroptosis in granulosa cells. J Ovarian Res. 2022;15(1):138.
Chi YN, Hai DM, Ma L, Cui YH, Hu HT, Liu N, et al. Protective effects of leonurine hydrochloride on pyroptosis in premature ovarian insufficiency via regulating NLRP3/GSDMD pathway. Int Immunopharmacol. 2023;114: 109520.
Chaudhary GR, Yadav PK, Yadav AK, Tiwari M, Gupta A, Sharma A, et al. Necrosis and necroptosis in germ cell depletion from mammalian ovary. J Cell Physiol. 2019;234(6):8019–27.
Li H, Jing Y, Qu X, Yang J, Pan P, Liu X, et al. The Activation of Reticulophagy by ER Stress through the ATF4-MAP1LC3A-CCPG1 Pathway in Ovarian Granulosa Cells Is Linked to Apoptosis and Necroptosis. Int J Mol Sci. 2023;24(3).
Delcour C, Amazit L, Patino LC, Magnin F, Fagart J, Delemer B, et al. ATG7 and ATG9A loss-of-function variants trigger autophagy impairment and ovarian failure. Genet Med. 2019;21(4):930–8.
Lu X, Bao H, Cui L, Zhu W, Zhang L, Xu Z, et al. hUMSC transplantation restores ovarian function in POI rats by inhibiting autophagy of theca-interstitial cells via the AMPK/mTOR signaling pathway. Stem Cell Res Ther. 2020;11(1):268.
Han S, Li H, Lu R, Feng J, Tang K, Li S, et al. Effect and mechanism of pearl on ovarian function of rats with premature ovarian failure induced by tripterygium glycosides. J Tradit Complement Med. 2023;13(4):368–78.
Zhang CR, Zhu WN, Tao W, Lin WQ, Cheng CC, Deng H, et al. Moxibustion against Cyclophosphamide-Induced Premature Ovarian Failure in Rats through Inhibiting NLRP3-/Caspase-1-/GSDMD-Dependent Pyroptosis. Evid Based Complement Alternat Med. 2021;2021:8874757.
Lambrinoudaki I, Paschou SA, Lumsden MA, Faubion S, Makrakis E, Kalantaridou S, et al. Premature ovarian insufficiency: A toolkit for the primary care physician. Maturitas. 2021;147:53–63.
Armeni E, Paschou SA, Goulis DG, Lambrinoudaki I. Hormone therapy regimens for managing the menopause and premature ovarian insufficiency. Best Pract Res Clin Endocrinol Metab. 2021;35(6): 101561.
McGlacken-Byrne SM, Conway GS. Premature ovarian insufficiency. Best Pract Res Clin Obstet Gynaecol. 2022;81:98–110.
Poggio F, Del Mastro L, Bruzzone M, Ceppi M, Razeti MG, Fregatti P, et al. Safety of systemic hormone replacement therapy in breast cancer survivors: a systematic review and meta-analysis. Breast Cancer Res Treat. 2022;191(2):269–75.
Collaborative Group on Hormonal Factors in Breast C. Type and timing of menopausal hormone therapy and breast cancer risk: individual participant meta-analysis of the worldwide epidemiological evidence. Lancet. 2019;394(10204):1159–68.
Ugras SK, Layeequr RR. Hormone replacement therapy after breast cancer: Yes, No or maybe? Mol Cell Endocrinol. 2021;525: 111180.
Cho HW, Ouh YT, Lee JK, Hong JH. Effects of hormone therapy on recurrence in endometrial cancer survivors: a nationwide study using the Korean Health Insurance Review and Assessment Service database. J Gynecol Oncol. 2019;30(4): e51.
Shao A, Tu S, Lu J, Zhang J. Crosstalk between stem cell and spinal cord injury: pathophysiology and treatment strategies. Stem Cell Res Ther. 2019;10(1):238.
Fu YX, Ji J, Shan F, Li J, Hu R. Human mesenchymal stem cell treatment of premature ovarian failure: new challenges and opportunities. Stem Cell Res Ther. 2021;12(1):161.
Huang Y, Zhu M, Liu Z, Hu R, Li F, Song Y, et al. Bone marrow mesenchymal stem cells in premature ovarian failure: Mechanisms and prospects. Front Immunol. 2022;13: 997808.
Qu Q, Liu L, Cui Y, Liu H, Yi J, Bing W, et al. miR-126-3p containing exosomes derived from human umbilical cord mesenchymal stem cells promote angiogenesis and attenuate ovarian granulosa cell apoptosis in a preclinical rat model of premature ovarian failure. Stem Cell Res Ther. 2022;13(1):352.
Ling L, Feng X, Wei T, Wang Y, Wang Y, Wang Z, et al. Human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation improves ovarian function in rats with premature ovarian insufficiency (POI) at least partly through a paracrine mechanism. Stem Cell Res Ther. 2019;10(1):46.
Yoon SY, Yoon JA, Park M, Shin EY, Jung S, Lee JE, et al. Recovery of ovarian function by human embryonic stem cell-derived mesenchymal stem cells in cisplatin-induced premature ovarian failure in mice. Stem Cell Res Ther. 2020;11(1):255.
Sun B, Ma Y, Wang F, Hu L, Sun Y. miR-644-5p carried by bone mesenchymal stem cell-derived exosomes targets regulation of p53 to inhibit ovarian granulosa cell apoptosis. Stem Cell Res Ther. 2019;10(1):360.
Li Z, Zhang M, Tian Y, Li Q, Huang X. Mesenchymal Stem Cells in Premature Ovarian Insufficiency: Mechanisms and Prospects. Front Cell Dev Biol. 2021;9: 718192.
Zhuang WZ, Lin YH, Su LJ, Wu MS, Jeng HY, Chang HC, et al. Mesenchymal stem/stromal cell-based therapy: mechanism, systemic safety and biodistribution for precision clinical applications. J Biomed Sci. 2021;28(1):28.
Jiang Y, Zhang P, Zhang X, Lv L, Zhou Y. Advances in mesenchymal stem cell transplantation for the treatment of osteoporosis. Cell Prolif. 2021;54(1): e12956.
Marodin MSJ, Godoy JA, Alves-Paiva RM, Alvarez K, Mitsugi TG, Krepischi ACV, et al. Preclinical Evaluation of the Tumorigenic and Immunomodulatory Properties of Human Bone Marrow Mesenchymal Stromal Cell Populations with Clonal Trisomy 5. Stem Cells Int. 2022;2022:1613636.
Csobonyeiova M, Beerova N, Klein M, Debreova-Cehakova M, Danisovic L. Cell-Based and Selected Cell-Free Therapies for Myocardial Infarction: How Do They Compare to the Current Treatment Options? Int J Mol Sci. 2022;23(18).
Guo YM, Sun TC, Wang HP, Chen X. Research progress of melatonin (MT) in improving ovarian function: a review of the current status. Aging (Albany NY). 2021;13(13):17930–47.
Feng J, Ma WW, Li HX, Pei XY, Deng SL, Jia H, et al. Melatonin prevents cyclophosphamide-induced primordial follicle loss by inhibiting ovarian granulosa cell apoptosis and maintaining AMH expression. Front Endocrinol (Lausanne). 2022;13: 895095.
Barberino RS, Lins T, Monte APO, Gouveia BB, Campinho DSP, Palheta RC Jr, et al. Melatonin Attenuates Cyclophosphamide-Induced Primordial Follicle Loss by Interaction with MT(1) Receptor and Modulation of PTEN/Akt/FOXO3a Proteins in the Mouse Ovary. Reprod Sci. 2022;29(9):2505–14.
Huang J, Shan W, Li N, Zhou B, Guo E, Xia M, et al. Melatonin provides protection against cisplatin-induced ovarian damage and loss of fertility in mice. Reprod Biomed Online. 2021;42(3):505–19.
Li M, Xiao YB, Wei L, Liu Q, Liu PY, Yao JF. Beneficial Effects of Traditional Chinese Medicine in the Treatment of Premature Ovarian Failure. Evid Based Complement Alternat Med. 2022;2022:5413504.
Cai L, Zong DK, Tong GQ, Li L. Apoptotic mechanism of premature ovarian failure and rescue effect of Traditional Chinese Medicine: a review. J Tradit Chin Med. 2021;41(3):492–8.
Tang LP, Gong XX, Chen QQ, Wang S, Chen XF, Zhou XL, et al. CiteSpace knowledge mapping of traditional Chinese medicine in treatment of premature ovarian failure. Zhongguo Zhong Yao Za Zhi. 2022;47(13):3658–66.
Li HF, Zhang JX, Chen WJ. Dissecting the efficacy of the use of acupuncture and Chinese herbal medicine for the treatment of premature ovarian insufficiency (POI): A systematic review and metaanalysis. Heliyon. 2023;9(10): e20498.
Wang L, Li M, Liu J, Nie G, Li Y, Yang H. Protective effect of Huyang Yangkun Formula on ovarian function in premature ovarian insufficiency rats based on apoptotic mechanism. J Ethnopharmacol. 2021;280: 114477.
Jiang XL, Tai H, Kuang JS, Zhang JY, Cui SC, Lu YX, et al. Jian-Pi-Yi-Shen decoction inhibits mitochondria-dependent granulosa cell apoptosis in a rat model of POF. Aging (Albany NY). 2022;14(20):8321–45.
Song Z, Song K, Zhao H, He Y, Hu J. Network analysis and experimental approach to investigate the potential therapeutic mechanism of zishen yutai pills on premature ovarian insufficiency. Heliyon. 2023;9(9): e20025.
Chen S, Lu Y, Chen Y, Xu J, Chen L, Zhao W, et al. The effect of Bu Shen Huo Xue Tang on autoimmune premature ovarian insufficiency via Modulation of the Nrf2/Keap1 signaling pathway in mice. J Ethnopharmacol. 2021;273: 113996.
Zhou F, Song Y, Liu X, Zhang C, Li F, Hu R, et al. Si-Wu-Tang facilitates ovarian function through improving ovarian microenvironment and angiogenesis in a mouse model of premature ovarian failure. J Ethnopharmacol. 2021;280: 114431.
Dou X, Jin X, Chen X, Zhou Q, Chen H, Wen M, et al. Bu-Shen-Ning-Xin decoction alleviates premature ovarian insufficiency (POI) by regulating autophagy of granule cells through activating PI3K/AKT/mTOR pathway. Gynecol Endocrinol. 2022;38(9):754–64.
Li N, Wang J, Wang X, Sun J, Li Z. Icariin exerts a protective effect against d-galactose induced premature ovarian failure via promoting DNA damage repair. Biomed Pharmacother. 2019;118: 109218.
Xiu Z, Tang S, Kong P, Yan M, Tong X, Liu X, et al. Zigui-Yichong-Fang protects against cyclophosphamide-induced premature ovarian insufficiency via the SIRT1/Foxo3a pathway. J Ethnopharmacol. 2023;314: 116608.
Fan W, Zhang Y, Wang D, Wang C, Yang J. The impact of Yiwei decoction on the LncRNA and CircRNA regulatory networks in premature ovarian insufficiency. Heliyon. 2023;9(9): e20022.
Liu X, Song Y, Zhou F, Zhang C, Li F, Hu R, et al. Network and experimental pharmacology on mechanism of Si-Wu-tang improving ovarian function in a mouse model of premature ovarian failure induced by cyclophosphamide. J Ethnopharmacol. 2023;301: 115842.
Xie L, Wu S, Cao D, Li M, Liu J, Nie G, et al. Huyang yangkun formula protects against 4-Vinylcyclohexene diepoxide-induced premature ovarian insufficiency in rats via the Hippo-JAK2/STAT3 signaling pathway. Biomed Pharmacother. 2019;116: 109008.
Chen Y, Fan X, Ma K, Wang K, Tian C, Li M, et al. Bushen Culuan Decoction Ameliorates Premature Ovarian Insufficiency by Acting on the Nrf2/ARE Signaling Pathway to Alleviate Oxidative Stress. Front Pharmacol. 2022;13: 857932.
Huang Y, Hu R, Liu Z, Geng Y, Li F, Song Y, et al. Bushen Huoxue recipe ameliorates ovarian function via promoting BMSCs proliferation and homing to ovaries in POI mice. Phytomedicine. 2024;129: 155630.
Li XF, Wang ZQ, Xu HY, Liu H, Sheng Y, Xu J, et al. Effects of Zishen Yutai Pills on in vitro Fertilization-Embryo Transfer Outcomes in Patients with Diminished Ovarian Reserve: A Prospective, Open-Labeled, Randomized and Controlled Study. Chin J Integr Med. 2023;29(4):291–8.
Jiang Y, Zhang Z, Cha L, Li L, Zhu D, Fang Z, et al. Resveratrol Plays a Protective Role against Premature Ovarian Failure and Prompts Female Germline Stem Cell Survival. Int J Mol Sci. 2019;20(14).
Herrero Y, Velazquez C, Pascuali N, May M, Abramovich D, Scotti L, et al. Resveratrol alleviates doxorubicin-induced damage in mice ovary. Chem Biol Interact. 2023;376: 110431.
Zheng S, Ma M, Chen Y, Li M. Effects of quercetin on ovarian function and regulation of the ovarian PI3K/Akt/FoxO3a signalling pathway and oxidative stress in a rat model of cyclophosphamide-induced premature ovarian failure. Basic Clin Pharmacol Toxicol. 2022;130(2):240–53.
Yan F, Zhao Q, Gao H, Wang X, Xu K, Wang Y, et al. Exploring the mechanism of (-)-Epicatechin on premature ovarian insufficiency based on network pharmacology and experimental evaluation. Biosci Rep. 2021;41(2).
Wu YY, Liang CY, Liu TT, Liang YM, Li SJ, Lu YY, et al. Protective roles and mechanisms of polysaccharides from Dendrobium officinal on natural aging-induced premature ovarian failure. Biomed Pharmacother. 2018;101:953–60.
Xin L, Li F, Yu H, Xiong Q, Hou Q, Meng Y. Honokiol alleviates radiation-induced premature ovarian failure via enhancing Nrf2. Am J Reprod Immunol. 2023;90(4): e13769.
Zheng H, Liang X, Zhou H, Zhou T, Liu X, Duan J, et al. 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.
Shang Z, Fan M, Zhang J, Wang Z, Jiang S, Li W. Red Ginseng Improves D-galactose-Induced Premature Ovarian Failure in Mice Based on Network Pharmacology. Int J Mol Sci. 2023;24(9).
Xu X, Qu Z, Qian H, Li Z, Sun X, Zhao X, et al. Ginsenoside Rg1 ameliorates reproductive function injury in C57BL/6J mice induced by di-N-butyl-phthalate. Environ Toxicol. 2021;36(5):789–99.
Chen W, Luo H, Zhong Z, Wei J, Wang Y. The safety of Chinese medicine: A systematic review of endogenous substances and exogenous residues. Phytomedicine. 2023;108: 154534.
Yang B, Xie Y, Guo M, Rosner MH, Yang H, Ronco C. Nephrotoxicity and Chinese Herbal Medicine. Clin J Am Soc Nephrol. 2018;13(10):1605–11.
Kilis-Pstrusinska K, Wiela-Hojenska A. Nephrotoxicity of Herbal Products in Europe-A Review of an Underestimated Problem. Int J Mol Sci. 2021;22(8).
Xiong Y, Li M, Sun P, Liang W, Hornbeck RG, Che X, et al. Market access for Chinese herbal medicinal products in Europe-A ten-year review of relevant products, policies, and challenges. Phytomedicine. 2022;103: 154237.
Oesch F, Oesch-Bartlomowicz B, Efferth T. Toxicity as prime selection criterion among SARS-active herbal medications. Phytomedicine. 2021;85: 153476.
Wang S, Lin S, Zhu M, Li C, Chen S, Pu L, et al. Acupuncture Reduces Apoptosis of Granulosa Cells in Rats with Premature Ovarian Failure Via Restoring the PI3K/Akt Signaling Pathway. Int J Mol Sci. 2019;20(24).
Zhang H, Qin F, Liu A, Sun Q, Wang Q, Xie S, et al. Electro-acupuncture attenuates the mice premature ovarian failure via mediating PI3K/AKT/mTOR pathway. Life Sci. 2019;217:169–75.
Yan J, Zhao JY, Yin LY, Yan XQ, Jin XF. Effect of acupuncture at “Zhibian” (BL 54) through “Shuidao” (ST 28) on the expression of apoptosis-related factors in rats with premature ovarian insufficiency based on oxidative stress. Zhongguo Zhen Jiu. 2023;43(4):454–60.
Fan S, Fang YG. Research progress of acupuncture for the improvement of ovarian reserve by regulating different signal pathways. Zhen Ci Yan Jiu. 2022;47(7):644–8.
Geng Z, Nie X, Ling L, Li B, Liu P, Yuan L, et al. Electroacupuncture May Inhibit Oxidative Stress of Premature Ovarian Failure Mice by Regulating Intestinal Microbiota. Oxid Med Cell Longev. 2022;2022:4362317.
Lin G, Liu X, Cong C, Chen S, Xu L. Clinical efficacy of acupuncture for diminished ovarian reserve: a systematic review and meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne). 2023;14:1136121.
Shen J, Li HX, Lu G, Yin YL, Cheng J, Xia YB, et al. Protective effect of moxibustion preconditioning in rats with premature ovarian insufficiency. Zhen Ci Yan Jiu. 2023;48(3):267–73.
Yi A, Qin X, Du Z, Wang T, Liu F. Clinical Observation on the Improvement of Serum Sex Hormone and Ovarian Function in Premature Ovarian Failure Patients with Deficiency-Cold Syndrome by Combining Wenjing Decoction with Tiaobu Chongren Acupuncture and Moxibustion. Evid Based Complement Alternat Med. 2021;2021:3926822.
Yin Y, Li H, Qin Y, Chen T, Zhang Z, Lu G, et al. Moxibustion mitigates mitochondrial dysfunction and NLRP3 inflammatory activation in cyclophosphamide-induced premature ovarian insufficiency rats. Life Sci. 2023;314: 121283.
Vo KCT, Kawamura K. In Vitro Activation Early Follicles: From the Basic Science to the Clinical Perspectives. Int J Mol Sci. 2021;22(7).
Zhai J, Zhang J, Zhang L, Liu X, Deng W, Wang H, et al. Autotransplantation of the ovarian cortex after in-vitro activation for infertility treatment: a shortened procedure. Hum Reprod. 2021;36(8):2134–47.
Huang QY, Chen SR, Chen JM, Shi QY, Lin S. Therapeutic options for premature ovarian insufficiency: an updated review. Reprod Biol Endocrinol. 2022;20(1):28.
Devos M, Grosbois J, Demeestere I. Interaction between PI3K/AKT and Hippo pathways during in vitro follicular activation and response to fragmentation and chemotherapy exposure using a mouse immature ovary model. Biol Reprod. 2020;102(3):717–29.
Tanaka Y, Hsueh AJ, Kawamura K. Surgical approaches of drug-free in vitro activation and laparoscopic ovarian incision to treat patients with ovarian infertility. Fertil Steril. 2020;114(6):1355–7.
Zhai J, Yao G, Dong F, Bu Z, Cheng Y, Sato Y, et al. In Vitro Activation of Follicles and Fresh Tissue Auto-transplantation in Primary Ovarian Insufficiency Patients. J Clin Endocrinol Metab. 2016;101(11):4405–12.
Zhang X, Han T, Yan L, Jiao X, Qin Y, Chen ZJ. Resumption of Ovarian Function After Ovarian Biopsy/Scratch in Patients With Premature Ovarian Insufficiency. Reprod Sci. 2019;26(2):207–13.
Lunding SA, Pors SE, Kristensen SG, Landersoe SK, Jeppesen JV, Flachs EM, et al. Biopsying, fragmentation and autotransplantation of fresh ovarian cortical tissue in infertile women with diminished ovarian reserve. Hum Reprod. 2019;34(10):1924–36.
Atkinson L, Martin F, Sturmey RG. Intraovarian injection of platelet-rich plasma in assisted reproduction: too much too soon? Hum Reprod. 2021;36(7):1737–50.
Sfakianoudis K, Simopoulou M, Grigoriadis S, Pantou A, Tsioulou P, Maziotis E, et al. Reactivating Ovarian Function through Autologous Platelet-Rich Plasma Intraovarian Infusion: Pilot Data on Premature Ovarian Insufficiency, Perimenopausal, Menopausal, and Poor Responder Women. J Clin Med. 2020;9(6).
Cakiroglu Y, Saltik A, Yuceturk A, Karaosmanoglu O, Kopuk SY, Scott RT, et al. Effects of intraovarian injection of autologous platelet rich plasma on ovarian reserve and IVF outcome parameters in women with primary ovarian insufficiency. Aging (Albany NY). 2020;12(11):10211–22.
Ahmadian S, Sheshpari S, Pazhang M, Bedate AM, Beheshti R, Abbasi MM, et al. Intra-ovarian injection of platelet-rich plasma into ovarian tissue promoted rejuvenation in the rat model of premature ovarian insufficiency and restored ovulation rate via angiogenesis modulation. Reprod Biol Endocrinol. 2020;18(1):78.
El Bakly W, Medhat M, Shafei M, Tash R, Elrefai M, Shoukry Y, et al. Optimized platelet rich plasma releasate (O-rPRP) repairs galactosemia-induced ovarian follicular loss in rats by activating mTOR signaling and inhibiting apoptosis. Heliyon. 2020;6(9): e05006.
Seckin S, Ramadan H, Mouanness M, Kohansieh M, Merhi Z. Ovarian response to intraovarian platelet-rich plasma (PRP) administration: hypotheses and potential mechanisms of action. J Assist Reprod Genet. 2022;39(1):37–61.
Urman B, Boza A, Balaban B. Platelet-rich plasma another add-on treatment getting out of hand? How can clinicians preserve the best interest of their patients? Hum Reprod. 2019;34(11):2099–103.
Khattak H, Malhas R, Craciunas L, Afifi Y, Amorim CA, Fishel S, et al. Fresh and cryopreserved ovarian tissue transplantation for preserving reproductive and endocrine function: a systematic review and individual patient data meta-analysis. Hum Reprod Update. 2022;28(3):400–16.
Hoekman EJ, Louwe LA, Rooijers M, van der Westerlaken LAJ, Klijn NF, Pilgram GSK, et al. Ovarian tissue cryopreservation: Low usage rates and high live-birth rate after transplantation. Acta Obstet Gynecol Scand. 2020;99(2):213–21.
Ma L, Li X, Li C, Chen P, Lan Y, Huang Y, et al. Association of Coenzyme Q10 with Premature Ovarian Insufficiency. Reprod Sci. 2023;30(5):1548–54.
Ghahremani-Nasab M, Ghanbari E, Jahanbani Y, Mehdizadeh A, Yousefi M. Premature ovarian failure and tissue engineering. J Cell Physiol. 2020;235(5):4217–26.
Shelling AN, Ahmed Nasef N. The Role of Lifestyle and Dietary Factors in the Development of Premature Ovarian Insufficiency. Antioxidants (Basel). 2023;12(8).
Ishizuka B. Current Understanding of the Etiology, Symptomatology, and Treatment Options in Premature Ovarian Insufficiency (POI). Front Endocrinol (Lausanne). 2021;12: 626924.
Guo Q, Liang XY. Advances in the genetic etiology of premature ovarian insufficiency. Sci China Life Sci. 2023;66(7):1705–7.
Identification of pathogenic DNA variants in premature ovarian insufficiency. Nat Med. 2023;29(2):315–6.
Gingrich J, Ticiani E, Veiga-Lopez A. Placenta Disrupted: Endocrine Disrupting Chemicals and Pregnancy. Trends Endocrinol Metab. 2020;31(7):508–24.
Quiros-Alcala L, Hansel NN, McCormack M, Calafat AM, Ye X, Peng RD, et al. Exposure to bisphenols and asthma morbidity among low-income urban children with asthma. J Allergy Clin Immunol. 2021;147(2):577–86 e7.
Liu W, Sun Y, Liu N, Hou J, Huo X, Zhao Y, et al. Indoor exposure to phthalates and its burden of disease in China. Indoor Air. 2022;32(4): e13030.
Pagoni A, Arvaniti OS, Kalantzi OI. Exposure to phthalates from personal care products: Urinary levels and predictors of exposure. Environ Res. 2022;212(Pt A): 113194.
Wei F, Mortimer M, Cheng H, Sang N, Guo LH. Parabens as chemicals of emerging concern in the environment and humans: A review. Sci Total Environ. 2021;778: 146150.
Chang X, Fang Y, Wang Y, Wang F, Shang L, Zhong R. Microplastic pollution in soils, plants, and animals: A review of distributions, effects and potential mechanisms. Sci Total Environ. 2022;850: 157857.
Huang Y, Xiao L, Li F, Xiao M, Lin D, Long X, et al. Microbial Degradation of Pesticide Residues and an Emphasis on the Degradation of Cypermethrin and 3-phenoxy Benzoic Acid: A Review. Molecules. 2018;23(9).
Geng Y, Liu Z, Hu R, Huang Y, Li F, Ma W, et al. Toxicity of microplastics and nanoplastics: invisible killers of female fertility and offspring health. Front Physiol. 2023;14:1254886.
Sharma P, Bilkhiwal N, Chaturvedi P, Kumar S, Khetarpal P. Potential environmental toxicant exposure, metabolizing gene variants and risk of PCOS-A systematic review. Reprod Toxicol. 2021;103:124–32.
Chianese R, Troisi J, Richards S, Scafuro M, Fasano S, Guida M, et al. Bisphenol A in Reproduction: Epigenetic Effects. Curr Med Chem. 2018;25(6):748–70.
Rebuzzini P, Fabozzi G, Cimadomo D, Ubaldi FM, Rienzi L, Zuccotti M, et al. Multi- and Transgenerational Effects of Environmental Toxicants on Mammalian Reproduction. Cells. 2022;11(19).
Acknowledgements
The work was supported by National Natural Science Foundation of China (No. 81904008 and No. 82274309). Thanks to BioRender. The figure is created with BioRender.com.
Author information
Authors and Affiliations
Contributions
All authors contributed to the conception and design of the review. Y.H. wrote the first draft of the manuscript. Y. H. and Z. L. extracted the information from the reviewed studies. All authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to particpate
The authors have no relevant financial or non-financial interests to disclose.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Huang, Y., Liu, Z., Geng, Y. et al. The risk factors, pathogenesis and treatment of premature ovarian insufficiency. J Ovarian Res 18, 134 (2025). https://doi.org/10.1186/s13048-025-01714-2
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s13048-025-01714-2




