Aging hallmarks of the primate ovary revealed by spatiotemporal transcriptomics

Huifen Lu, Ying Jing, Chen Zhang, Shuai Ma, Weiqi Zhang, Daoyuan Huang, Bin Zhang, Yuesheng Zuo, Yingying Qin, Guang-Hui Liu, Yang Yu, Jing Qu, Si Wang

PDF(44093 KB)
PDF(44093 KB)
Protein Cell ›› 2024, Vol. 15 ›› Issue (5) : 364-384. DOI: 10.1093/procel/pwad063
RESEARCH ARTICLE

Aging hallmarks of the primate ovary revealed by spatiotemporal transcriptomics

Author information +
History +

Abstract

The ovary is indispensable for female reproduction, and its age-dependent functional decline is the primary cause of infertility. However, the molecular basis of ovarian aging in higher vertebrates remains poorly understood. Herein, we apply spatiotemporal transcriptomics to benchmark architecture organization as well as cellular and molecular determinants in young primate ovaries and compare these to aged primate ovaries. From a global view, somatic cells within the non-follicle region undergo more pronounced transcriptional fluctuation relative to those in the follicle region, likely constituting a hostile microenvironment that facilitates ovarian aging. Further, we uncovered that inflammation, the senescent-associated secretory phenotype, senescence, and fibrosis are the likely primary contributors to ovarian aging (PCOA). Of note, we identified spatial co-localization between a PCOA-featured spot and an unappreciated MT2 (Metallothionein 2) highly expressing spot (MT2high) characterized by high levels of inflammation, potentially serving as an aging hotspot in the primate ovary. Moreover, with advanced age, a subpopulation of MT2high accumulates, likely disseminating and amplifying the senescent signal outward. Our study establishes the first primate spatiotemporal transcriptomic atlas, advancing our understanding of mechanistic determinants underpinning primate ovarian aging and unraveling potential biomarkers and therapeutic targets for aging and age-associated human ovarian disorders.

Keywords

spatial transcriptome / primate / ovary / aging / senescence / inflammation

Cite this article

Download citation ▾
Huifen Lu, Ying Jing, Chen Zhang, Shuai Ma, Weiqi Zhang, Daoyuan Huang, Bin Zhang, Yuesheng Zuo, Yingying Qin, Guang-Hui Liu, Yang Yu, Jing Qu, Si Wang. Aging hallmarks of the primate ovary revealed by spatiotemporal transcriptomics. Protein Cell, 2024, 15(5): 364‒384 https://doi.org/10.1093/procel/pwad063

References

[1]
Adashi EY. Endocrinology of the ovary. Hum Reprod 1994; 9:815–27.
CrossRef Google scholar
[2]
Aging Atlas C. Aging Atlas: a multi-omics database for aging biology. Nucleic Acids Res 2021; 49:D825–30.
CrossRef Google scholar
[3]
Aging Biomarker C, Bao H, Cao J et al. Biomarkers of aging. Sci China Life Sci 2023; 66:893–1066.
CrossRef Google scholar
[4]
Ahmed TA, Ahmed SM, El-Gammal Z et al. Oocyte aging: the role of cellular and environmental factors and impact on female fertility. Adv Exp Med Biol 2020; 1247:109–23.
CrossRef Google scholar
[5]
Amargant F, Manuel SL, Tu Q et al. Ovarian stiffness increases with age in the mammalian ovary and depends on collagen and hyaluronan matrices. Aging Cell 2020; 19:e13259.
CrossRef Google scholar
[6]
Ata B, Seyhan A, Seli E. Diminished ovarian reserve versus ovarian aging: overlaps and differences. Curr Opin Obstet Gynecol 2019; 31:139–47.
CrossRef Google scholar
[7]
Auersperg N, Wong AS, Choi KC et al. Ovarian surface epithelium: biology, endocrinology, and pathology. Endocr Rev 2001; 22:255–88.
CrossRef Google scholar
[8]
Bai H, Mu L, Qiu L et al. Complement C3 regulates inflammatory response and monocyte/macrophage phagocytosis of Streptococcus agalactiae in a teleost fish. Int J Mol Sci 2022; 23:15586.
CrossRef Google scholar
[9]
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:e74915.
CrossRef Google scholar
[10]
Birch J, Gil J. Senescence and the SASP: many therapeutic avenues. Genes Dev 2020; 34:1565–76.
CrossRef Google scholar
[11]
Broekmans FJ, Soules MR, Fauser BC. Ovarian aging: mechanisms and clinical consequences. Endocr Rev 2009; 30:465–93.
CrossRef Google scholar
[12]
Brown HM, Russell DL. Blood and lymphatic vasculature in the ovary: development, function and disease. Hum Reprod Update 2014; 20:29–39.
CrossRef Google scholar
[13]
Burris-Hiday SD, Scott EE. Steroidogenic cytochrome P450 17A1 structure and function. Mol Cell Endocrinol 2021; 528:111261.
CrossRef Google scholar
[14]
Cai Y, Song W, Li J et al. The landscape of aging. Sci China Life Sci 2022; 65:2354–454.
CrossRef Google scholar
[15]
Castrillon DH, Quade BJ, Wang TY et al. The human VASA gene is specifically expressed in the germ cell lineage. Proc Natl Acad Sci U S A 2000; 97:9585–90.
CrossRef Google scholar
[16]
Cheng C, Liu ZG. Autophagy and the metabolism of misfolding protein. Adv Exp Med Biol 2019; 1206:375–420.
CrossRef Google scholar
[17]
Cheng S, Li Z, Gao R et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell 2021; 184:792–809e23.
CrossRef Google scholar
[18]
Debacq-Chainiaux F, Erusalimsky JD, Campisi J et al. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat Protoc 2009; 4:1798–806.
CrossRef Google scholar
[19]
Dries R, Zhu Q, Dong R et al. Giotto: a toolbox for integrative analysis and visualization of spatial expression data. Genome Biol 2021; 22:78.
CrossRef Google scholar
[20]
Duffy DM, Ko C, Jo M et al. Ovulation: parallels with inflammatory processes. Endocr Rev 2019; 40:369–416.
CrossRef Google scholar
[21]
Efremova M, Vento-Tormo M, Teichmann SA et al. CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat Protoc 2020; 15:1484–506.
CrossRef Google scholar
[22]
Fan X, Chuva de Sousa Lopes SM. Molecular makeup of the human adult ovary. Curr Opin Endocr Metab Res 2021; 18:187–93.
CrossRef Google scholar
[23]
He S, Wang LH, Liu Y et al. Single-cell transcriptome profiling of an adult human cell atlas of 15 major organs. Genome Biol 2020a; 21:294.
CrossRef Google scholar
[24]
He X, Memczak S, Qu J et al. Single-cell omics in ageing: a young and growing field. Nat Metab 2020b; 2:293–302.
CrossRef Google scholar
[25]
Huang D, Zuo Y, Zhang C et al. A single-nucleus transcriptomic atlas of primate testicular aging reveals exhaustion of the spermatogonial stem cell reservoir and loss of Sertoli cell homeostasis. Protein Cell 2022; 14:887-907.
CrossRef Google scholar
[26]
Husain MA, Laurent B, Plourde M. APOE and Alzheimer’s disease: from lipid transport to physiopathology and therapeutics. Front Neurosci 2021; 15:630502.
CrossRef Google scholar
[27]
Isola JVV, Ocañas SR, Hubbart CR et al. A single-cell atlas of the aging murine ovary. bioRxiv 2023.
CrossRef Google scholar
[28]
Jia L, Wang W, Liang J et al. Analyzing the cellular and molecular atlas of ovarian mesenchymal cells provides a strategy against female reproductive aging. Sci China Life Sci 2023; 66:2818–2836.
CrossRef Google scholar
[29]
Jing Y, Zuo Y, Yu Y et al. Single-nucleus profiling unveils a geroprotective role of the FOXO3 in primate skeletal muscle aging. Protein Cell 2023; 14:497–512.
CrossRef Google scholar
[30]
Kanehisa M, Furumichi M, Sato Y et al. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res 2021; 49:D545–51.
CrossRef Google scholar
[31]
Komatsu K, Masubuchi S. Observation of the dynamics of follicular development in the ovary. Reprod Med Biol 2017; 16:21–7.
CrossRef Google scholar
[32]
Lehallier B, Gate D, Schaum N et al. Undulating changes in human plasma proteome profiles across the lifespan. Nat Med 2019; 25:1843–50.
CrossRef Google scholar
[33]
Leng SX, Pawelec G. Single-cell immune atlas for human aging and frailty. Life Med 2022; 1:67–70.
CrossRef Google scholar
[34]
Lengyel E, Li Y, Weigert M et al. A molecular atlas of the human postmenopausal fallopian tube and ovary from single-cell RNA and ATAC sequencing. Cell Rep 2022; 41:111838.
CrossRef Google scholar
[35]
Li J, Zheng Y, Yan P et al. A single-cell transcriptomic atlas of primate pancreatic islet aging. Natl Sci Rev 2021; 8:nwaa127.
CrossRef Google scholar
[36]
Liberzon A, Birger C, Thorvaldsdóttir H et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 2015; 1:417–25.
CrossRef Google scholar
[37]
Lin SN, Musso A, Wang J et al. Human intestinal myofibroblasts deposited collagen VI enhances adhesiveness for T cells—a novel mechanism for maintenance of intestinal inflammation. Matrix Biol 2022; 113:1–21.
CrossRef Google scholar
[38]
Liu J, Sun D, Liu J et al. FibroAtlas: a database for the exploration of fibrotic diseases and their genes. Cardiol Res Pract 2019; 2019:4237285.
CrossRef Google scholar
[39]
López-Otín C, Blasco MA, Partridge L et al. Hallmarks of aging: an expanding universe. Cell 2023; 186:243–78.
CrossRef Google scholar
[40]
Lovell TM, Gladwell RT, Groome NP et al. Ovarian follicle development in the laying hen is accompanied by divergent changes in inhibin A, inhibin B, activin A and follistatin production in granulosa and theca layers. J Endocrinol 2003; 177:45–55.
CrossRef Google scholar
[41]
Ma S, Sun S, Geng L et al. Caloric restriction reprograms the single-cell transcriptional landscape of Rattus Norvegicus aging. Cell 2020; 180:984–1001.e22.
CrossRef Google scholar
[42]
Ma S, Sun S, Li J et al. Single-cell transcriptomic atlas of primate cardiopulmonary aging. Cell Res 2021; 31:415–32.
CrossRef Google scholar
[43]
Ma S, Chi X, Cai Y et al. Decoding aging hallmarks at the single- cell level. Annu Rev Biomed Data Sci 2023; 6:129–52.
CrossRef Google scholar
[44]
Machlin JH, Shikanov A. Single-cell RNA-sequencing of retrieved human oocytes and eggs in clinical practice and for human ovarian cell atlasing. Mol Reprod Dev 2022; 89:597–607.
CrossRef Google scholar
[45]
Mara JN, Zhou LT, Larmore M et al. Ovulation and ovarian wound healing are impaired with advanced reproductive age. Aging (Albany NY) 2020; 12:9686–713.
CrossRef Google scholar
[46]
Mishina T, Tabata N, Hayashi T et al. Single-oocyte transcriptome analysis reveals aging-associated effects influenced by life stage and calorie restriction. Aging Cell 2021; 20:e13428.
CrossRef Google scholar
[47]
Pal L, Santoro N. Premature ovarian failure (POF): discordance between somatic and reproductive aging. Ageing Res Rev 2002; 1:413–23.
CrossRef Google scholar
[48]
Perry JR, Murray A, Day FR et al. Molecular insights into the aetiology of female reproductive ageing. Nat Rev Endocrinol 2015; 11:725–34.
CrossRef Google scholar
[49]
Petrosino JM, Disilvestro D, Ziouzenkova O. Aldehyde dehydrogenase 1A1: friend or foe to female metabolism? Nutrients 2014; 6:950–73.
CrossRef Google scholar
[50]
Read A, Schröder M. The unfolded protein response: an overview. Biology (Basel) 2021; 10:384.
CrossRef Google scholar
[51]
Rouillard AD, Gundersen GW, Fernandez NF et al. The harmonizome: a collection of processed datasets gathered to serve and mine knowledge about genes and proteins. Database (Oxford) 2016; 2016:baw100.
CrossRef Google scholar
[52]
Sasano H, Suzuki T. Localization of steroidogenesis and steroid receptors in human corpus luteum classification of human corpus luteum (CL) into estrogen-producing degenerating CL, and nonsteroid-producing degenerating CL. Semin Reprod Endocrinol 1997; 15:345–51.
CrossRef Google scholar
[53]
Shi Y, Guo Y, Zhou J et al. A spatiotemporal gene expression and cell atlases of the developing rat ovary. Cell Prolif 2023; 56:e13516.
CrossRef Google scholar
[54]
Sun Y, Li Q, Kirkland JL. Targeting senescent cells for a healthier longevity: the roadmap for an era of global aging. Life Med 2022; 1:103–19.
CrossRef Google scholar
[55]
Sun G, Zheng Y, Fu X et al. Single-cell transcriptomic atlas of mouse cochlear aging. Protein Cell 2023; 14:180–201.
CrossRef Google scholar
[56]
Tang F, Barbacioru C, Wang Y et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods 2009; 6:377–82.
CrossRef Google scholar
[57]
Tew WP. Ovarian cancer in the older woman. J Geriatr Oncol 2016; 7:354–61.
CrossRef Google scholar
[58]
Trapnell C, Cacchiarelli D, Grimsby J et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 2014; 32:381–6.
CrossRef Google scholar
[59]
Vollenhoven B, Hunt S. Ovarian ageing and the impact on female fertility. F1000Res 2018; 7:F1000 Faculty Rev-1835.
CrossRef Google scholar
[60]
Wagner M, Yoshihara M, Douagi I et al. Single-cell analysis of human ovarian cortex identifies distinct cell populations but no oogonial stem cells. Nat Commun 2020; 11:1147.
CrossRef Google scholar
[61]
Wang S, Hu B, Ding Z et al. ATF6 safeguards organelle homeostasis and cellular aging in human mesenchymal stem cells. Cell Discov 2018; 4:2.
CrossRef Google scholar
[62]
Wang S, Zheng Y, Li J et al. Single-cell transcriptomic atlas of primate ovarian aging. Cell 2020; 180:585–600 e519.
CrossRef Google scholar
[63]
Wang S, Cheng F, Ji Q et al. Hyperthermia differentially affects specific human stem cells and their differentiated derivatives. Protein Cell 2022; 13:615–22.
CrossRef Google scholar
[64]
Wei Y, Yu R, Cheng S et al. Single-cell profiling of mouse and primate ovaries identifies high levels of EGFR for stromal cells in ovarian aging. Mol Ther Nucleic Acids 2023; 31:1–12.
CrossRef Google scholar
[65]
Woad KJ, Robinson RS. Luteal angiogenesis and its control. Theriogenology 2016; 86:221–8.
CrossRef Google scholar
[66]
Wu T, Hu E, Xu S et al. clusterProfiler 40: a universal enrichment tool for interpreting omics data. Innovation (Camb) 2021; 2:100141.
CrossRef Google scholar
[67]
Wu J, Liu Y, Song Y et al. Aging conundrum: a perspective for ovarian aging. Front Endocrinol (Lausanne) 2022; 13:952471.
CrossRef Google scholar
[68]
Yuan L, Yin P, Yan H et al. Single-cell transcriptome analysis of human oocyte ageing. J Cell Mol Med 2021; 25:6289–303.
CrossRef Google scholar
[69]
Zhang W, Zhang S, Yan P et al. A single-cell transcriptomic landscape of primate arterial aging. Nat Commun 2020; 11:2202.
CrossRef Google scholar
[70]
Zhang B, Yan H, Liu X et al. SenoIndex: S100A8/S100A9 as a novel aging biomarker. Life Med 2023; 2:lnad022.
CrossRef Google scholar
[71]
Zhao H, Ji Q, Wu Z et al. Destabilizing heterochromatin by APOE mediates senescence. Nat Aging 2022; 2:303–16.
CrossRef Google scholar
[72]
Zhou Y, Zhou B, Pache L et al. Metascape provides a biologist- oriented resource for the analysis of systemslevel datasets. Nat Commun 2019; 10:1523.
CrossRef Google scholar
[73]
Zhou T, Kiran M, Lui KO et al. Decoding liver fibrogenesis with single-cell technologies. Life Med 2022; 1:333–44.
CrossRef Google scholar
[74]
Zhu Z, Xu W, Liu L. Ovarian aging: mechanisms and intervention strategies. Med Rev (Berlin, Germany) 2022; 2:590–610.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2023 The Author(s) 2023. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(44093 KB)

Accesses

Citations

Detail

Sections
Recommended

/