Exploration of the mechanism and therapy of ovarian aging by targeting cellular senescence

Weicheng Tang , Kaichen Wang , Yourong Feng , Kuan-Hao Tsui , Keshav K. Singh , Michael B. Stout , Shixuan Wang , Meng Wu

Life Medicine ›› 2025, Vol. 4 ›› Issue (1) : lnaf004

PDF (1511KB)
Life Medicine ›› 2025, Vol. 4 ›› Issue (1) : lnaf004 DOI: 10.1093/lifemedi/lnaf004
Review

Exploration of the mechanism and therapy of ovarian aging by targeting cellular senescence

Author information +
History +
PDF (1511KB)

Abstract

The ovary is a crucial gonadal organ that supports female reproductive and endocrine functions. Ovarian aging can result in decreased fertility and dysfunction across multiple organs. Research has demonstrated that cellular senescence in various cell types within the ovary can trigger a decline in ovarian function through distinct stress responses, resulting in ovarian aging. This review explores how cellular senescence may contribute to ovarian aging and reproductive failure. Additionally, we discuss the factors that cause ovarian cellular senescence, including the accumulation of advanced glycation end products, oxidative stress, mitochondrial dysfunction, DNA damage, telomere shortening, and exposure to chemotherapy. Furthermore, we discuss senescence in six distinct cell types, including oocytes, granulosa cells, ovarian theca cells, immune cells, ovarian surface epithelium, and ovarian endothelial cells, inside the ovary and explore their contribution to the accelerated ovarian aging. Lastly, we describe potential senotherapeutics for the treatment of ovarian aging and offer novel strategies for ovarian longevity.

Keywords

ovarian aging / cellular senescence / oocyte / granulosa cells / senotherapy

Cite this article

Download citation ▾
Weicheng Tang, Kaichen Wang, Yourong Feng, Kuan-Hao Tsui, Keshav K. Singh, Michael B. Stout, Shixuan Wang, Meng Wu. Exploration of the mechanism and therapy of ovarian aging by targeting cellular senescence. Life Medicine, 2025, 4(1): lnaf004 DOI:10.1093/lifemedi/lnaf004

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Ata B , Seli E . Strategies for controlled ovarian stimulation in the setting of ovarian aging. Semin Reprod Med 2015; 33: 436- 48.

[2]

Minkin MJ . Menopause: hormones, lifestyle, and optimizing aging. Obstet Gynecol Clin North Am 2019; 46: 501- 14.

[3]

Couzin-Frankel J . Reproductive biology. Faulty DNA repair linked to ovarian aging in mice and humans. Science 2013; 339: 749.

[4]

Hong W , Wang B , Zhu Y , et al. Female germline stem cells: aging and anti-aging. J Ovarian Res 2022; 15: 79.

[5]

Sang Q , Ray PF , Wang L . Understanding the genetics of human infertility. Science 2023; 380: 158- 63.

[6]

Ogrodnik M , Salmonowicz H , Jurk D , et al. Expansion and cell-cycle arrest: common denominators of cellular senescence. Trends Biochem Sci 2019; 44: 996- 1008.

[7]

van Deursen JM . The role of senescent cells in ageing. Nature 2014; 509: 439- 46.

[8]

Secomandi L , Borghesan M , Velarde M , et al. The role of cellular senescence in female reproductive aging and the potential for senotherapeutic interventions. Hum Reprod Update 2022; 28: 172- 89.

[9]

Lopez-Otin C , Blasco MA , Partridge L , et al. The hallmarks of aging. Cell 2013; 153: 1194- 217.

[10]

Armanios M . The role of telomeres in human disease. Annu Rev Genomics Hum Genet 2022; 23: 363- 81.

[11]

Nassrally MS , Lau A , Wise K , et al. Cell cycle arrest in replicative senescence is not an immediate consequence of telomere dysfunction. Mech Ageing Dev 2019; 179: 11- 22.

[12]

Kinugawa C , Murakami T , Okamura K , et al. Telomerase activity in normal ovaries and premature ovarian failure. Tohoku J Exp Med 2000; 190: 231- 8.

[13]

Werner CM , Hecksteden A , Morsch A , et al. Differential effects of endurance, interval, and resistance training on telomerase activity and telomere length in a randomized, controlled study. Eur Heart J 2019; 40: 34- 46.

[14]

Uysal F , Kosebent EG , Toru HS , et al. Decreased expression of tert and telomeric proteins as human ovaries age may cause telomere shortening. J Assist Reprod Genet 2021; 38: 429- 41.

[15]

Kosebent EG , Ozturk S . Telomere associated gene expression as well as tert protein level and telomerase activity are altered in the ovarian follicles of aged mice. Sci Rep 2021; 11: 15569.

[16]

Ozturk S . The close relationship between oocyte aging and telomere shortening, and possible interventions for telomere protection. Mech Ageing Dev 2024; 218: 111913.

[17]

Liu L , Blasco M , Trimarchi J , et al. An essential role for functional telomeres in mouse germ cells during fertilization and early development. Dev Biol 2002; 249: 74- 84.

[18]

Kosebent EG , Uysal F , Ozturk S . The altered expression of telomerase components and telomere-linked proteins may associate with ovarian aging in mouse. Exp Gerontol 2020; 138: 110975.

[19]

Robinson LG Jr , Pimentel R , Wang F , et al. Impaired reproductive function and fertility preservation in a woman with a dyskeratosis congenita. J Assist Reprod Genet 2020; 37: 1221- 5.

[20]

Cheng EH , Chen SU , Lee TH , et al. Evaluation of telomere length in cumulus cells as a potential biomarker of oocyte and embryo quality. Hum Reprod 2013; 28: 929- 36.

[21]

Toupance S , Fattet AJ , Thornton SN , et al. Ovarian telomerase and female fertility. Biomedicines 2021; 9: 842.

[22]

Xu X , Chen X , Zhang X , et al. Impaired telomere length and telomerase activity in peripheral blood leukocytes and granulosa cells in patients with biochemical primary ovarian insufficiency. Hum Reprod 2017; 32: 201- 7.

[23]

Miranda-Furtado CL , Luchiari HR , Chielli Pedroso DC , et al. Skewed x-chromosome inactivation and shorter telomeres associate with idiopathic premature ovarian insufficiency. Fertil Steril 2018; 110: 476- 85.e1.

[24]

Polonio AM , Medrano M , Chico-Sordo L , et al. Impaired telomere pathway and fertility in senescence-accelerated mice prone 8 females with reproductive senescence. Aging (Albany NY) 2023; 15: 4600- 24.

[25]

Jackson SP , Bartek J . The DNA-damage response in human biology and disease. Nature 2009; 461: 1071- 8.

[26]

Shreeya T , Ansari MS , Kumar P , et al. Senescence: a DNA damage response and its role in aging and neurodegenerative diseases. Front Aging 2023; 4: 1292053.

[27]

Soleimani R , Heytens E , Darzynkiewicz Z , et al. Mechanisms of chemotherapy-induced human ovarian aging: double strand DNA breaks and microvascular compromise. Aging (Albany NY) 2011; 3: 782- 93.

[28]

Johnson J , Keefe DL . Ovarian aging: breaking up is hard to fix. Sci Transl Med 2013; 5: 172fs5.

[29]

Ruth KS , Day FR , Hussain J , et al; Biobank-based Integrative Omics Study (BIOS) Consortium. Genetic insights into biological mechanisms governing human ovarian ageing. Nature 2021; 596: 393- 7.

[30]

Turan V , Oktay K . Brca-related atm-mediated DNA double-strand break repair and ovarian aging. Hum Reprod Update 2020; 26: 43- 57.

[31]

Zhang T , Ren T , Lin H , et al. Ash1l contributes to oocyte apoptosis by regulating DNA damage. Am J Physiol Cell Physiol 2022; 323: C1264- 73.

[32]

Zhang D , Zhang X , Zeng M , et al. Increased DNA damage and repair deficiency in granulosa cells are associated with ovarian aging in rhesus monkey. J Assist Reprod Genet 2015; 32: 1069- 78.

[33]

Sha C , Chen L , Lin L , et al. Trdmt1 participates in the DNA damage repair of granulosa cells in premature ovarian failure. Aging (Albany NY) 2021; 13: 15193- 213.

[34]

Liguori I , Russo G , Curcio F , et al. Oxidative stress, aging, and diseases. Clin Interv Aging 2018; 13: 757- 72.

[35]

Zhang H , Davies KJA , Forman HJ . Oxidative stress response and nrf2 signaling in aging. Free Radic Biol Med 2015; 88: 314- 36.

[36]

Mahdi H , Tahereh H , Esmaiel S , et al. Vitamins e and c prevent apoptosis of testicular and ovarian tissues following mancozeb exposure in the first-generation mouse pups. Toxicol Ind Health 2019; 35: 136- 44.

[37]

Lim J , Luderer U . Oxidative damage increases and antioxidant gene expression decreases with aging in the mouse ovary. Biol Reprod 2011; 84: 775- 82.

[38]

Yang L , Chen Y , Liu Y , et al. The role of oxidative stress and natural antioxidants in ovarian aging. Front Pharmacol 2020; 11: 617843.

[39]

Shaeib F , Banerjee J , Maitra D , et al. Impact of hydrogen peroxide-driven fenton reaction on mouse oocyte quality. Free Radic Biol Med 2013; 58: 154- 9.

[40]

Chen Q , Gao L , Li J , et al. Alpha-ketoglutarate improves meiotic maturation of porcine oocytes and promotes the development of pa embryos, potentially by reducing oxidative stress through the nrf2 pathway. Oxid Med Cell Longev 2022; 2022: 7113793.

[41]

Ekoue DN , He C , Diamond AM , et al. Manganese superoxide dismutase and glutathione peroxidase-1 contribute to the rise and fall of mitochondrial reactive oxygen species which drive oncogenesis. Biochim Biophys Acta Bioenerg 2017; 1858: 628- 32.

[42]

Wang S , Zheng Y , Li J , et al. Single-cell transcriptomic atlas of primate ovarian aging. Cell 2020; 180: 585- 600.e19.

[43]

Yang H , Qazi IH , Pan B , et al. Dietary selenium supplementation ameliorates female reproductive efficiency in aging mice. Antioxidants (Basel) 2019; 8: 634.

[44]

Katz-Jaffe MG , Lane SL , Parks JC , et al. Antioxidant intervention attenuates aging-related changes in the murine ovary and oocyte. Life (Basel) 2020; 10: 250.

[45]

Li F , Wang Y , Xu M , 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.

[46]

Wang L , Tang J , Wang L , et al. Oxidative stress in oocyte aging and female reproduction. J Cell Physiol 2021; 236: 7966- 83.

[47]

Lin X , Dai Y , Tong X , et al. Excessive oxidative stress in cumulus granulosa cells induced cell senescence contributes to endometriosis-associated infertility. Redox Biol 2020; 30: 101431.

[48]

Zhang Z , Zhang L , Zhou L , et al. Redox signaling and unfolded protein response coordinate cell fate decisions under er stress. Redox Biol 2019; 25: 101047.

[49]

Tatone C , Carbone MC , Falone S , et al. Age-dependent changes in the expression of superoxide dismutases and catalase are associated with ultrastructural modifications in human granulosa cells. Mol Hum Reprod 2006; 12: 655- 60.

[50]

Qian Y , Shao L , Yuan C , et al. Implication of differential peroxiredoxin 4 expression with age in ovaries of mouse and human for ovarian aging. Curr Mol Med 2016; 16: 243- 51.

[51]

Qiao J , Wang ZB , Feng HL , et al. The root of reduced fertility in aged women and possible therapentic options: current status and future perspects. Mol Aspects Med 2014; 38: 54- 85.

[52]

Sohel MMH , Akyuz B , Konca Y , et al. Oxidative stress modulates the expression of apoptosis-associated micrornas in bovine granulosa cells in vitro. Cell Tissue Res 2019; 376: 295- 308.

[53]

Martini H , Passos JF . Cellular senescence: all roads lead to mitochondria. FEBS J 2023; 290: 1186- 202.

[54]

May-Panloup P , Boucret L , Chao de la Barca JM , et al. Ovarian ageing: the role of mitochondria in oocytes and follicles. Hum Reprod Update 2016; 22: 725- 43.

[55]

Chow J , Rahman J , Achermann JC , et al. Mitochondrial disease and endocrine dysfunction. Nat Rev Endocrinol 2017; 13: 92- 104.

[56]

Tiosano D , Mears JA , Buchner DA . Mitochondrial dysfunction in primary ovarian insufficiency. Endocrinology 2019; 160: 2353- 66.

[57]

Chiang JL , Shukla P , Pagidas K , et al. Mitochondria in ovarian aging and reproductive longevity. Ageing Res Rev 2020; 63: 101168.

[58]

Miwa S , Kashyap S , Chini E , et al. Mitochondrial dysfunction in cell senescence and aging. J Clin Invest 2022; 132: e158447.

[59]

Lu X , Liu Y , Xu J , et al. Mitochondrial dysfunction in cumulus cells is related to decreased reproductive capacity in advanced-age women. Fertil Steril 2022; 118: 393- 404.

[60]

Shiiba I , Takeda K , Nagashima S , et al. Overview of mitochondrial e3 ubiquitin ligase mitol/march5 from molecular mechanisms to diseases. Int J Mol Sci 2020; 21: 3781.

[61]

Czajkowska K , Ajduk A . Mitochondrial activity and redox status in oocytes from old mice: the interplay between maternal and postovulatory aging. Theriogenology 2023; 204: 18- 30.

[62]

Lee S , Jeong SY , Lim WC , et al. Mitochondrial fission and fusion mediators, hfis1 and opa1, modulate cellular senescence. J Biol Chem 2007; 282: 22977- 83.

[63]

Ramirez-Camacho I , Flores-Herrera O , Zazueta C . The relevance of the supramolecular arrangements of the respiratory chain complexes in human diseases and aging. Mitochondrion 2019; 47: 266- 72.

[64]

Lin L , Gao W , Chen Y , et al. Reactive oxygen species-induced siah1 promotes granulosa cells’ senescence in premature ovarian failure. J Cell Mol Med 2022; 26: 2417- 27.

[65]

Nagata S , Tatematsu K , Kansaku K , et al. Effect of aging on mitochondria and metabolism of bovine granulosa cells. J Reprod Dev 2020; 66: 547- 54.

[66]

Duran HE , Simsek-Duran F , Oehninger SC , et al. The association of reproductive senescence with mitochondrial quantity, function, and DNA integrity in human oocytes at different stages of maturation. Fertil Steril 2011; 96: 384- 8.

[67]

Van Blerkom J . Mitochondrial function in the human oocyte and embryo and their role in developmental competence. Mitochondrion 2011; 11: 797- 813.

[68]

Iwata H , Goto H , Tanaka H , et al. Effect of maternal age on mitochondrial DNA copy number, atp content and ivf outcome of bovine oocytes. Reprod Fertil Dev 2011; 23: 424- 32.

[69]

Murakoshi Y , Sueoka K , Takahashi K , et al. Embryo developmental capability and pregnancy outcome are related to the mitochondrial DNA copy number and ooplasmic volume. J Assist Reprod Genet 2013; 30: 1367- 75.

[70]

Shi YQ , Zhu XT , Zhang SN , et al. Premature ovarian insufficiency: a review on the role of oxidative stress and the application of antioxidants. Front Endocrinol (Lausanne) 2023; 14: 1172481.

[71]

Yang L , Lin X , Tang H , et al. Mitochondrial DNA mutation exacerbates female reproductive aging via impairment of the nadh/nad(+) redox. Aging Cell 2020; 19: e13206.

[72]

Chan CC , Liu VW , Lau EY , et al. Mitochondrial DNA content and 4977 bp deletion in unfertilized oocytes. Mol Hum Reprod 2005; 11: 843- 6.

[73]

Fragouli E , Wells D . Mitochondrial DNA assessment to determine oocyte and embryo viability. Semin Reprod Med 2015; 33: 401- 9.

[74]

Ju W , Zhao Y , Yu Y , et al. Mechanisms of mitochondrial dysfunction in ovarian aging and potential interventions. Front Endocrinol (Lausanne) 2024; 15: 1361289.

[75]

Vlassara H , Striker GE . Advanced glycation endproducts in diabetes and diabetic complications. Endocrinol Metab Clin North Am 2013; 42: 697- 719.

[76]

Sell DR , Monnier VM . Structure elucidation of a senescence cross-link from human extracellular matrix. Implication of pentoses in the aging process. J Biol Chem 1989; 264: 21597- 602.

[77]

Tatone C , Amicarelli F , Carbone MC , et al. Cellular and molecular aspects of ovarian follicle ageing. Hum Reprod Update 2008; 14: 131- 42.

[78]

Pertynska-Marczewska M , Diamanti-Kandarakis E . Aging ovary and the role for advanced glycation end products. Menopause 2017; 24: 345- 51.

[79]

Coughlan MT , Cooper ME , Forbes JM . Renal microvascular injury in diabetes: rage and redox signaling. Antioxid Redox Signal 2007; 9: 331- 42.

[80]

Stensen MH , Tanbo T , Storeng R , et al. Advanced glycation end products and their receptor contribute to ovarian ageing. Hum Reprod 2014; 29: 125- 34.

[81]

Takahashi N , Harada M , Azhary JMK , et al. Accumulation of advanced glycation end products in follicles is associated with poor oocyte developmental competence. Mol Hum Reprod 2019; 25: 684- 94.

[82]

Kandaraki EA , Chatzigeorgiou A , Papageorgiou E , et al. Advanced glycation end products interfere in luteinizing hormone and follicle stimulating hormone signaling in human granulosa kgn cells. Exp Biol Med (Maywood) 2018; 243: 29- 33.

[83]

Mouanness M , Merhi Z . Impact of dietary advanced glycation end products on female reproduction: review of potential mechanistic pathways. Nutrients 2022; 14: 966.

[84]

Tatone C , Heizenrieder T , Di Emidio G , et al. Evidence that carbonyl stress by methylglyoxal exposure induces DNA damage and spindle aberrations, affects mitochondrial integrity in mammalian oocytes and contributes to oocyte ageing. Hum Reprod 2011; 26: 1843- 59.

[85]

Ott C , Jacobs K , Haucke E , et al. Role of advanced glycation end products in cellular signaling. Redox Biol 2014; 2: 411- 29.

[86]

Leonardis D , Basta G , Mallamaci F , et al. Circulating soluble receptor for advanced glycation end product (srage) and left ventricular hypertrophy in patients with chronic kidney disease (ckd). Nutr Metab Cardiovasc Dis 2012; 22: 748- 55.

[87]

Fujii EY , Nakayama M . The measurements of rage, vegf, and ages in the plasma and follicular fluid of reproductive women: the influence of aging. Fertil Steril 2010; 94: 694- 700.

[88]

Tatone C , Eichenlaub-Ritter U , Amicarelli F . Dicarbonyl stress and glyoxalases in ovarian function. Biochem Soc Trans 2014; 42: 433- 8.

[89]

Diamanti-Kandarakis E , Piperi C , Patsouris E , et al. Immunohistochemical localization of advanced glycation end-products (ages) and their receptor (rage) in polycystic and normal ovaries. Histochem Cell Biol 2007; 127: 581- 9.

[90]

Harlow CR , Rae M , Davidson L , et al. Lysyl oxidase gene expression and enzyme activity in the rat ovary: regulation by follicle-stimulating hormone, androgen, and transforming growth factor-beta superfamily members in vitro. Endocrinology 2003; 144: 154- 62.

[91]

Levi N , Papismadov N , Solomonov I , et al. The ecm path of senescence in aging: components and modifiers. FEBS J 2020; 287: 2636- 46.

[92]

Wang Z , Liu H , Xu C . Cellular senescence in the treatment of ovarian cancer. Int J Gynecol Cancer 2018; 28: 895- 902.

[93]

Xiong J , Xue L , Li Y , et al. Therapy of endocrine disease: novel protection and treatment strategies for chemotherapy-associated ovarian damage. Eur J Endocrinol 2021; 184: R177- 92.

[94]

Blumenfeld Z . Chemotherapy and fertility. Best Pract Res Clin Obstet Gynaecol 2012; 26: 379- 90.

[95]

Piasecka-Srader J , Blanco FF , Delman DH , et al. Tamoxifen prevents apoptosis and follicle loss from cyclophosphamide in cultured rat ovaries. Biol Reprod 2015; 92: 132.

[96]

Xu Z , Takahashi N , Harada M , et al. The role of cellular senescence in cyclophosphamide-induced primary ovarian insufficiency. Int J Mol Sci 2023; 24: 17193.

[97]

Luan Y , Edmonds ME , Woodruff TK , et al. Inhibitors of apoptosis protect the ovarian reserve from cyclophosphamide. J Endocrinol 2019; 240: 243- 56.

[98]

Jiang Y , Zhang Z , Cha L , et al. Resveratrol plays a protective role against premature ovarian failure and prompts female germline stem cell survival. Int J Mol Sci 2019; 20: 3605.

[99]

Iqubal A , Iqubal MK , Sharma S , et al. Molecular mechanism involved in cyclophosphamide-induced cardiotoxicity: old drug with a new vision. Life Sci 2019; 218: 112- 31.

[100]

Luo Q , Yin N , Zhang L , et al. Role of sdf-1/cxcr4 and cytokines in the development of ovary injury in chemotherapy drug induced premature ovarian failure mice. Life Sci 2017; 179: 103- 9.

[101]

Spears N , Lopes F , Stefansdottir A , et al. Ovarian damage from chemotherapy and current approaches to its protection. Hum Reprod Update 2019; 25: 673- 93.

[102]

Morgan S , Anderson RA , Gourley C , et al. How do chemotherapeutic agents damage the ovary? Hum Reprod Update 2012; 18: 525- 35.

[103]

Tuppi M , Kehrloesser S , Coutandin DW , et al. Oocyte DNA damage quality control requires consecutive interplay of chk2 and ck1 to activate p63. Nat Struct Mol Biol 2018; 25: 261- 9.

[104]

Ma P , Xiao H , Yu C , et al. Enhanced cisplatin chemotherapy by iron oxide nanocarrier-mediated generation of highly toxic reactive oxygen species. Nano Lett 2017; 17: 928- 37.

[105]

Ayres LS , Berger M , Durli I , et al. Kallikrein-kinin system and oxidative stress in cisplatin-induced ovarian toxicity. Reprod Toxicol 2020; 93: 1- 9.

[106]

Eldani M , Luan Y , Xu PC , et al. Continuous treatment with cisplatin induces the oocyte death of primordial follicles without activation. FASEB J 2020; 34: 13885- 99.

[107]

Ben-Aharon I , Bar-Joseph H , Tzarfaty G , et al. Doxorubicin-induced ovarian toxicity. Reprod Biol Endocrinol 2010; 8: 20.

[108]

Demeestere I , Racape J , Dechene J , et al. Gonadal function recovery in patients with advanced hodgkin lymphoma treated with a petadapted regimen: prospective analysis of a randomized phase iii trial (ahl2011). J Clin Oncol 2021; 39: 3251- 60.

[109]

Gao Y , Wu T , Tang X , et al. Increased cellular senescence in doxorubicin-induced murine ovarian injury: effect of senolytics. Geroscience 2023; 45: 1775- 90.

[110]

Wang Y , Liu M , Johnson SB , et al. Doxorubicin obliterates mouse ovarian reserve through both primordial follicle atresia and overactivation. Toxicol Appl Pharmacol 2019; 381: 114714.

[111]

Niringiyumukiza JD , Cai H , Chen L , et al. Protective properties of glycogen synthase kinase-3 inhibition against doxorubicin-induced oxidative damage to mouse ovarian reserve. Biomed Pharmacother 2019; 116: 108963.

[112]

Tokarska-Schlattner M , Zaugg M , Zuppinger C , et al. New insights into doxorubicin-induced cardiotoxicity: the critical role of cellular energetics. J Mol Cell Cardiol 2006; 41: 389- 405.

[113]

Wenningmann N , Knapp M , Ande A , et al. Insights into doxorubicin-induced cardiotoxicity: molecular mechanisms, preventive strategies, and early monitoring.Mol Pharmacol 2019; 96: 219- 32.

[114]

Luu AZ , Chowdhury B , Al-Omran M , et al. Role of endothelium in doxorubicin-induced cardiomyopathy. JACC Basic Transl Sci 2018; 3: 861- 70.

[115]

Wang JJ , Ge W , Zhai QY , et al. Single-cell transcriptome landscape of ovarian cells during primordial follicle assembly in mice. PLoS Biol 2020; 18: e3001025.

[116]

Fan X , Bialecka M , Moustakas I , et al. Single-cell reconstruction of follicular remodeling in the human adult ovary. Nat Commun 2019; 10: 3164.

[117]

Sauer MV , Paulson RJ , Lobo RA . A preliminary report on oocyte donation extending reproductive potential to women over 40. N Engl J Med 1990; 323: 1157- 60.

[118]

te Velde ER , Pearson PL . The variability of female reproductive ageing. Hum Reprod Update 2002; 8: 141- 54.

[119]

Wright VC , Schieve LA , Reynolds MA , et al; Division of Reproductive Health, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention (CDC). Assisted reproductive technology surveillance--united states, 2002. MMWR Surveill Summ 2005; 54: 1- 24.

[120]

Baumann K . Keeping oocytes young. Nat Rev Mol Cell Biol 2021; 22: 586.

[121]

Eftekhari Moghadam AR , Saki G , Hemadi M , et al. Effect of dehy-droepiandrosterone on meiotic spindle structure and oocyte quality in mice. Iran J Basic Med Sci 2018; 21: 1020- 5.

[122]

Moghadam ARE , Moghadam MT , Hemadi M , et al. Oocyte quality and aging. JBRA Assist Reprod 2022; 26: 105- 22.

[123]

Pasquariello R , Ermisch AF , Silva E , et al. Alterations in oocyte mitochondrial number and function are related to spindle defects and occur with maternal aging in mice and humansdagger. Biol Reprod 2019; 100: 971- 81.

[124]

Hamatani T , Falco G , Carter MG , et al. Age-associated alteration of gene expression patterns in mouse oocytes.Hum Mol Genet 2004; 13: 2263- 78.

[125]

Liang X , Ma J , Schatten H , et al. Epigenetic changes associated with oocyte aging. Sci China Life Sci 2012; 55: 670- 6.

[126]

Yan H , Miranda EAD , Jin S , et al. Primary oocytes with cellular senescence features are involved in ovarian aging in mice. Sci Rep 2024; 14: 13606.

[127]

Alberico HC , Woods DC . Role of granulosa cells in the aging ovarian landscape: a focus on mitochondrial and metabolic function. Front Physiol 2021; 12: 800739.

[128]

Clarke H . Control of mammalian oocyte development by interactions with the maternal follicular environment. Results Probl Cell Differ 2017; 63: 17- 41.

[129]

Su YQ , Sugiura K , Eppig JJ . Mouse oocyte control of granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med 2009; 27: 32- 42.

[130]

Guo J , Zhang T , Guo Y , et al. Oocyte stage-specific effects of mtor determine granulosa cell fate and oocyte quality in mice. Proc Natl Acad Sci U S A 2018; 115: E5326- 33.

[131]

Matsuda F , Inoue N , Manabe N , et al. Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. J Reprod Dev 2012; 58: 44- 50.

[132]

Devesa J , Caicedo D . The role of growth hormone on ovarian functioning and ovarian angiogenesis. Front Endocrinol (Lausanne) 2019; 10: 450.

[133]

Lin N , Lin J , Plosch T , et al. An oxidative stress-related gene signature in granulosa cells is associated with ovarian aging. Oxid Med Cell Longev 2022; 2022: 1070968.

[134]

Abdelnour SA , Swelum AA , Abd El-Hack ME , et al. Cellular and functional adaptation to thermal stress in ovarian granulosa cells in mammals. J Therm Biol 2020; 92: 102688.

[135]

Faraonio R . Oxidative stress and cell senescence process. Antioxidants (Basel) 2022; 11: 1718.

[136]

Kobayashi H , Yoshimoto C , Matsubara S , et al. Altered energy metabolism, mitochondrial dysfunction, and redox imbalance influencing reproductive performance in granulosa cells and oocyte during aging. Reprod Sci 2023; 31: 906- 16.

[137]

Reeves G . Specific stroma in the cortex and medulla of the ovary. Cell types and vascular supply in relation to follicular apparatus and ovulation. Obstet Gynecol 1971; 37: 832- 44.

[138]

Rotgers E , Jorgensen A , Yao HH . At the crossroads of fate-somatic cell lineage specification in the fetal gonad. Endocr Rev 2018; 39: 739- 59.

[139]

Young JM , McNeilly AS . Theca: the forgotten cell of the ovarian follicle. Reproduction 2010; 140: 489- 504.

[140]

Ethun KF , Wood CE , Parker CR Jr , et al. Effect of ovarian aging on androgen biosynthesis in a cynomolgus macaque model. Climacteric 2012; 15: 82- 92.

[141]

Shen L , Liu J , Luo A , et al. The stromal microenvironment and ovarian aging: mechanisms and therapeutic opportunities. J Ovarian Res 2023; 16: 237.

[142]

Shen L , Chen Y , Cheng J , et al. Ccl5 secreted by senescent theca-interstitial cells inhibits preantral follicular development via granulosa cellular apoptosis. J Cell Physiol 2019; 234: 22554- 64.

[143]

Tajima K , Orisaka M , Hosokawa K , et al. Effects of ovarian theca cells on apoptosis and proliferation of granulosa cells: changes during bovine follicular maturation. Biol Reprod 2002; 66: 1635- 9.

[144]

Kinnear HM , Tomaszewski CE , Chang FL , et al. The ovarian stroma as a new frontier. Reproduction 2020; 160: R25- 39.

[145]

Ben Yaakov T , Wasserman T , Aknin E , et al. Single-cell analysis of the aged ovarian immune system reveals a shift towards adaptive immunity and attenuated cell function. Elife 2023; 12: 74915.

[146]

Wu R , Van der Hoek KH , Ryan NK , et al. Macrophage contributions to ovarian function. Hum Reprod Update 2004; 10: 119- 33.

[147]

Tingen CM , Kiesewetter SE , Jozefik J , et al. A macrophage and theca cell-enriched stromal cell population influences growth and survival of immature murine follicles in vitro. Reproduction 2011; 141: 809- 20.

[148]

Turner EC , Hughes J , Wilson H , et al. Conditional ablation of macrophages disrupts ovarian vasculature.Reproduction 2011; 141: 821- 31.

[149]

Cohen-Fredarow A , Tadmor A , Raz T , et al. Ovarian dendritic cells act as a double-edged pro-ovulatory and anti-inflammatory sword. Mol Endocrinol 2014; 28: 1039- 54.

[150]

Ma L , Lu H , Chen R , et al. Identification of key genes and potential new biomarkers for ovarian aging: a study based on rna-sequencing data. Front Genet 2020; 11: 590660.

[151]

Isola JVV , Ocanas SR , Hubbart CR , et al. A single-cell atlas of the aging mouse ovary. Nat Aging 2024; 4: 145- 62.

[152]

Jasti S , Warren BD , McGinnis LK , et al. The autoimmune regulator prevents premature reproductive senescence in female mice. Biol Reprod 2012; 86: 110.

[153]

Song P , An J , Zou MH . Immune clearance of senescent cells to combat ageing and chronic diseases. Cells 2020; 9: 671.

[154]

Coppe JP , Desprez PY , Krtolica A , et al. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 2010; 5: 99- 118.

[155]

Urzua U , Chacon C , Espinoza R , et al. Parity-dependent hemosiderin and lipofuscin accumulation in the reproductively aged mouse ovary. Anal Cell Pathol (Amst) 2018; 2018: 1289103.

[156]

Umehara T , Winstanley YE , Andreas E , et al. Female reproductive life span is extended by targeted removal of fibrotic collagen from the mouse ovary. Sci Adv 2022; 8: eabn4564.

[157]

Nicosia SV , Johnson JH . Surface morphology of ovarian mesothelium (surface epithelium) and of other pelvic and extrapelvic mesothelial sites in the rabbit. Int J Gynecol Pathol 1984; 3: 249- 60.

[158]

Osterholzer HO , Streibel EJ , Nicosia SV . Growth effects of protein hormones on cultured rabbit ovarian surface epithelial cells. Biol Reprod 1985; 33: 247- 58.

[159]

Kim KY , Park DW , Jeung EB , et al. Conditional knockout of brca1/2 and p53 in mouse ovarian surface epithelium: do they play a role in ovarian carcinogenesis? J Vet Sci 2010; 11: 291- 7.

[160]

Okamura H , Katabuchi H , Nitta M , et al. Structural changes and cell properties of human ovarian surface epithelium in ovarian patho-physiology. Microsc Res Tech 2006; 69: 469- 81.

[161]

Clow OL , Hurst PR , Fleming JS . Changes in the mouse ovarian surface epithelium with age and ovulation number. Mol Cell Endocrinol 2002; 191: 105- 11.

[162]

Wong AS , Leung PC . Role of endocrine and growth factors on the ovarian surface epithelium. J Obstet Gynaecol Res 2007; 33: 3- 16.

[163]

Vasickova K , Moran L , Gurin D , et al. Alleviation of endoplasmic reticulum stress by tauroursodeoxycholic acid delays senescence of mouse ovarian surface epithelium. Cell Tissue Res 2018; 374: 643- 52.

[164]

Laszczynska M , Brodowska A , Starczewski A , et al. Human post-menopausal ovary--hormonally inactive fibrous connective tissue or more? Histol Histopathol 2008; 23: 219- 26.

[165]

Zhu R , Ji X , Wu X , et al. Melatonin antagonizes ovarian aging via ythdf2-mapk-nf-kappab pathway.Genes & diseases 2022; 9: 494- 509.

[166]

Brown HM , Russell DL . Blood and lymphatic vasculature in the ovary: development, function and disease.Hum Reprod Update 2014; 20: 29- 39.

[167]

Brown HM , Robker RL , Russell DL . Development and hormonal regulation of the ovarian lymphatic vasculature. Endocrinology 2010; 151: 5446- 55.

[168]

Wu M , Tang W , Chen Y . Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of FOXP1. Nat Aging 2024; 4: 527- 45.

[169]

Di Micco R , Krizhanovsky V , Baker D , et al. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol 2021; 22: 75- 95.

[170]

Hense JD , Garcia DN , Isola JV , et al. Senolytic treatment reverses obesity-mediated senescent cell accumulation in the ovary. Geroscience 2022; 44: 1747- 59.

[171]

Du D , Tang X , Li Y , et al. Senotherapy protects against cisplatin-induced ovarian injury by removing senescent cells and alleviating DNA damage. Oxid Med Cell Longev 2022; 2022: 9144644.

[172]

Dou X , Sun Y , Li J , et al. Short-term rapamycin treatment increases ovarian lifespan in young and middle-aged female mice. Aging Cell 2017; 16: 825- 36.

[173]

Chen X , Tang Z , Guan H , et al. Rapamycin maintains the primordial follicle pool and protects ovarian reserve against cyclophosphamide-induced damage. J Reprod Dev 2022; 68: 287- 94.

[174]

Zhou L , Xie Y , Li S , et al. Rapamycin prevents cyclophosphamide-induced over-activation of primordial follicle pool through pi3k/akt/mtor signaling pathway in vivo. J Ovarian Res 2017; 10: 56.

[175]

Qin X , Du D , Chen Q , et al. Metformin prevents murine ovarian aging. Aging (Albany NY) 2019; 11: 3785- 94.

[176]

Zhang J , Ma X , Li Y , et al. Metformin intervention against ovarian toxicity during chemotherapy for early breast cancer: study protocol for a randomized double-blind placebo-controlled trial. Maturitas 2020; 137: 1- 6.

[177]

McCloskey CW , Cook DP , Kelly BS , et al. Metformin abrogates age-associated ovarian fibrosis. Clin Cancer Res 2020; 26: 632- 42.

[178]

Okamoto N , Sato Y , Kawagoe Y , et al. Short-term resveratrol treatment restored the quality of oocytes in aging mice. Aging (Albany NY) 2022; 14: 5628- 40.

[179]

Wu H , Xue J , Di H , et al. Resveratrol improves ovarian function in aged rat by inhibiting oxidative stress and activating the sirt1. Gen Physiol Biophys 2022; 41: 53- 61.

[180]

Zhu H , Li X , Qiao M , et al. Resveratrol alleviates inflammation and er stress through sirt1/nrf2 to delay ovarian aging in a short-lived fish. J Gerontol A Biol Sci Med Sci 2023; 78: 596- 602.

[181]

Yang C , Liu Q , Chen Y , et al. Melatonin delays ovarian aging in mice by slowing down the exhaustion of ovarian reserve. Commun Biol 2021; 4: 534.

[182]

Tamura H , Kawamoto M , Sato S , et al. Long-term melatonin treatment delays ovarian aging. J Pineal Res 2017; 62: e12381.

[183]

Hao EY , Chen H , Wang DH , et al. Melatonin regulates the ovarian function and enhances follicle growth in aging laying hens via activating the mammalian target of rapamycin pathway. Poult Sci 2020; 99: 2185- 95.

[184]

Zhang M , ShiYang X , Zhang Y , et al. Coenzyme q10 ameliorates the quality of postovulatory aged oocytes by suppressing DNA damage and apoptosis. Free Radic Biol Med 2019; 143: 84- 94.

[185]

Xu Y , Nisenblat V , Lu C , et al. Pretreatment with coenzyme q10 improves ovarian response and embryo quality in low-prognosis young women with decreased ovarian reserve: a randomized controlled trial. Reprod Biol Endocrinol 2018; 16: 29.

[186]

Ma L , Cai L , Hu M , et al. Coenzyme q10 supplementation of human oocyte in vitro maturation reduces postmeiotic aneuploidies. Fertil Steril 2020; 114: 331- 7.

[187]

Lindauer M , Hochhaus A . Dasatinib. Recent Results Cancer Res 2018; 212: 29- 68.

[188]

Hosseini A , Razavi BM , Banach M , et al. Quercetin and metabolic syndrome: a review. Phytother Res 2021; 35: 5352- 64.

[189]

Novais EJ , Tran VA , Johnston SN , et al. Long-term treatment with senolytic drugs dasatinib and quercetin ameliorates age-dependent intervertebral disc degeneration in mice. Nat Commun 2021; 12: 5213.

[190]

Xu M , Pirtskhalava T , Farr JN , et al. Senolytics improve physical function and increase lifespan in old age. Nat Med 2018; 24: 1246- 56.

[191]

Justice JN , Nambiar AM , Tchkonia T , et al. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine 2019; 40: 554- 63.

[192]

Hickson LJ , Langhi Prata LGP , Bobart SA , et al. Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of dasatinib plus quercetin in individuals with diabetic kidney disease. EBioMedicine 2019; 47: 446- 56.

[193]

Islam MT , Tuday E , Allen S , et al. Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue inflammation, and ameliorate metabolic function in old age. Aging Cell 2023; 22: e13767.

[194]

Cavalcante MB , Saccon TD , Nunes ADC , et al. Dasatinib plus quercetin prevents uterine age-related dysfunction and fibrosis in mice. Aging (Albany NY) 2020; 12: 2711- 22.

[195]

Wu M , Tang W , Chen Y , et al. Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of foxp1. Nat Aging 2024; 4: 527- 45.

[196]

Blagosklonny MV . Rapamycin for longevity: opinion article. Aging (Albany NY) 2019; 11: 8048- 67.

[197]

Selvarani R , Mohammed S , Richardson A . Effect of rapamycin on aging and age-related diseases-past and future. Geroscience 2021; 43: 1135- 58.

[198]

Lamming DW , Ye L , Sabatini DM , et al. Rapalogs and mtor inhibitors as anti-aging therapeutics. J Clin Invest 2013; 123: 980- 9.

[199]

Bjedov I , Rallis C . The target of rapamycin signalling pathway in ageing and lifespan regulation. Genes (Basel) 2020; 11: 1043.

[200]

Wang R , Yu Z , Sunchu B , et al. Rapamycin inhibits the secretory phenotype of senescent cells by a nrf2-independent mechanism. Aging Cell 2017; 16: 564- 74.

[201]

Garcia DN , Saccon TD , Pradiee J , et al. Effect of caloric restriction and rapamycin on ovarian aging in mice. Geroscience 2019; 41: 395- 408.

[202]

Foretz M , Guigas B , Bertrand L , et al. Metformin: from mechanisms of action to therapies. Cell Metab 2014; 20: 953- 66.

[203]

Martin-Montalvo A , Mercken EM , Mitchell SJ , et al. Metformin improves healthspan and lifespan in mice. Nat Commun 2013; 4: 2192.

[204]

Kulkarni AS , Gubbi S , Barzilai N . Benefits of metformin in attenuating the hallmarks of aging. Cell Metab 2020; 32: 15- 30.

[205]

Smieszek A , Strek Z , Kornicka K , et al. Antioxidant and anti-senescence effect of metformin on mouse olfactory ensheathing cells (moecs) may be associated with increased brain-derived neurotrophic factor levels-an ex vivo study. Int J Mol Sci 2017; 18: 872.

[206]

Meng X , Zhou J , Zhao CN , et al. Health benefits and molecular mechanisms of resveratrol: a narrative review. Foods 2020; 9: 340.

[207]

Zhang LX , Li CX , Kakar MU , et al. Resveratrol (rv): a pharmacological review and call for further research. Biomed Pharmacother 2021; 143: 112164.

[208]

Csiszar A , Sosnowska D , Wang M , et al. Age-associated proinflam-matory secretory phenotype in vascular smooth muscle cells from the non-human primate macaca mulatta: reversal by resveratrol treatment. J Gerontol A Biol Sci Med Sci 2012; 67: 811- 20.

[209]

Zhang L , Pitcher LE , Prahalad V , et al. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J 2023; 290: 1362- 83.

[210]

Wu M , Ma L , Xue L , et al. Resveratrol alleviates chemotherapy-induced oogonial stem cell apoptosis and ovarian aging in mice. Aging (Albany NY) 2019; 11: 1030- 44.

[211]

Jiki Z , Lecour S , Nduhirabandi F . Cardiovascular benefits of dietary melatonin: a myth or a reality? Front Physiol 2018; 9: 528.

[212]

Acuna-Castroviejo D , Escames G , Venegas C , et al. Extrapineal melatonin: sources, regulation, and potential functions. Cell Mol Life Sci 2014; 71: 2997- 3025.

[213]

Gomes PRL , Motta-Teixeira LC , Gallo CC , et al. Maternal pineal melatonin in gestation and lactation physiology, and in fetal development and programming. Gen Comp Endocrinol 2021; 300: 113633.

[214]

Zhang J , Chen Q , Du D , et al. Can ovarian aging be delayed by pharmacological strategies? Aging (Albany NY) 2019; 11: 817- 32.

[215]

Galano A , Tan DX , Reiter RJ . Melatonin: a versatile protector against oxidative DNA damage. Molecules 2018; 23: 530.

[216]

Tan DX , Hardeland R , Manchester LC , et al. The changing biological roles of melatonin during evolution: from an antioxidant to signals of darkness, sexual selection and fitness. Biol Rev Camb Philos Soc 2010; 85: 607- 23.

[217]

Carrasco C , Marchena AM , Holguin-Arevalo MS , et al. Anti-inflammatory effects of melatonin in a rat model of caerulein-induced acute pancreatitis. Cell Biochem Funct 2013; 31: 585- 90.

[218]

Yang Y , Cheung HH , Zhang C , et al. Melatonin as potential targets for delaying ovarian aging. Curr Drug Targets 2019; 20: 16- 28.

[219]

Nishihara T , Hashimoto S , Ito K , et al. Oral melatonin supplementation improves oocyte and embryo quality in women undergoing in vitro fertilization-embryo transfer. Gynecol Endocrinol 2014; 30: 359- 62.

[220]

Ben-Meir A , Kim K , McQuaid R , et al. Co-enzyme q10 supplementation rescues cumulus cells dysfunction in a maternal aging model. Antioxidants (Basel) 2019; 8: 58.

[221]

Rodriguez-Varela C , Labarta E . Does coenzyme q10 supplementation improve human oocyte quality? Int J Mol Sci 2021; 22: 9541.

[222]

Akarsu S , Gode F , Isik AZ , et al. The association between coenzyme q10 concentrations in follicular fluid with embryo morphokinetics and pregnancy rate in assisted reproductive techniques. J Assist Reprod Genet 2017; 34: 599- 605.

[223]

Zhou XY , Yang YZ , Zhang J , et al. Elevated cell-free mitochondria DNA level of patients with premature ovarian insufficiency. BMC Pregnancy Childbirth 2023; 23: 462.

[224]

Abu-Halima M , Becker LS , Ayesh BM , et al. Characterization of microrna in women with different ovarian reserve. Sci Rep 2021; 11: 13351.

[225]

Al Rashid K , Taylor A , Lumsden MA , et al. Association of the functional ovarian reserve with serum metabolomic profiling by nuclear magnetic resonance spectroscopy: a cross-sectional study of ~ 400 women. BMC Med 2020; 18: 247.

[226]

Yan S , Jin W , Ding J , et al. Machine-intelligence for developing a potent signature to predict ovarian response to tailor assisted reproduction technology. Aging (Albany NY) 2021; 13: 17137- 54.

[227]

Sonigo C , Jankowski S , Yoo O , et al. High-throughput ovarian follicle counting by an innovative deep learning approach.Sci Rep 2018; 8: 13499.

[228]

Henarejos-Castillo I , Aleman A , Martinez-Montoro B , et al. Machine learning-based approach highlights the use of a genomic variant profile for precision medicine in ovarian failure. J Pers Med 2021; 11: 609.

[229]

Ding T , Ren W , Wang T , et al. Assessment and quantification of ovarian reserve on the basis of machine learning models. Front Endocrinol (Lausanne) 2023; 14: 1087429.

RIGHTS & PERMISSIONS

The Author(s). Published by Oxford University Press on behalf of Higher Education Press.

AI Summary AI Mindmap
PDF (1511KB)

590

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/