Dynamic transcriptomic and regulatory networks underpinning the transition from fetal primordial germ cells to spermatogonia in mice

Jiexiang Zhao , Kang Tang , Gurong Jiang , Xinyan Yang , Manman Cui , Cong Wan , Zhaoxiang Ouyang , Yi Zheng , Zhaoting Liu , Mei Wang , Xiao-Yang Zhao , Gang Chang

Cell Proliferation ›› 2025, Vol. 58 ›› Issue (2) : e13755

PDF
Cell Proliferation ›› 2025, Vol. 58 ›› Issue (2) : e13755 DOI: 10.1002/cpr.13755
ORIGINAL ARTICLE

Dynamic transcriptomic and regulatory networks underpinning the transition from fetal primordial germ cells to spermatogonia in mice

Author information +
History +
PDF

Abstract

The transition from fetal primordial germ cells (PGCs) to spermatogonia (SPG) is critical for male germ cell development; however, the detailed transcriptomic dynamics and regulation underlying this transition remain poorly understood. Here by interrogating the comprehensive transcriptome atlas dataset of mouse male germ cells and gonadal cells development, we elucidated the regulatory networks underlying this transition. Our single-cell transcriptome analysis revealed that the transition from PGCs to SPG was characterized by global hypertranscription. A total of 315 highly active regulators were identified to be potentially involved in this transition, among which a non-transcription factor (TF) regulator TAGLN2 was validated to be essential for spermatogonial stem cells (SSCs) maintenance and differentiation. Metabolism profiling analysis also revealed dynamic changes in metabolism-related gene expression during PGC to SPG transition. Furthermore, we uncovered that intricate cell–cell communication exerted potential functions in the regulation of hypertranscription in germ cells by collaborating with stage-specific active regulators. Collectively, our work extends the understanding of molecular mechanisms underlying male germ cell development, offering insights into the recapitulation of germ cell generation in vitro.

Cite this article

Download citation ▾
Jiexiang Zhao, Kang Tang, Gurong Jiang, Xinyan Yang, Manman Cui, Cong Wan, Zhaoxiang Ouyang, Yi Zheng, Zhaoting Liu, Mei Wang, Xiao-Yang Zhao, Gang Chang. Dynamic transcriptomic and regulatory networks underpinning the transition from fetal primordial germ cells to spermatogonia in mice. Cell Proliferation, 2025, 58(2): e13755 DOI:10.1002/cpr.13755

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

IshikuraY, OhtaH, SatoT, et al. In vitro reconstitution of the whole male germ-cell development from mouse pluripotent stem cells. Cell Stem Cell. 2021;28:2167-2179.

[2]

HutchinsAP, YangZ, LiY, et al. Models of global gene expression define major domains of cell type and tissue identity. Nucleic Acids Res. 2017;45(5):2354-2367.

[3]

WangM, LiuX, ChangG, et al. Single-cell RNA sequencing analysis reveals sequential cell fate transition during human spermatogenesis. Cell Stem Cell. 2018;23(4):599-614.

[4]

Garcia-AlonsoL, Lorenzi V, MazzeoCI, et al. Single-cell roadmap of human gonadal development. Nature. 2022;607:540-547.

[5]

SalehiN, Totonchi M. The construction of a testis transcriptional cell atlas from embryo to adult reveals various somatic cells and their molecular roles. J Transl Med. 2023;21(1):859.

[6]

DongF, PingP, MaY, ChenXF. Application of single-cell RNA sequencing on human testicular samples: a comprehensive review. Int J Biol Sci. 2023;19(7):2167-2197.

[7]

LiL, DongJ, YanL, et al. Single-cell RNA-Seq analysis maps development of human germline cells and gonadal niche interactions. Cell Stem Cell. 2017;20(6):858-873.

[8]

GuoJ, GrowEJ, MlcochovaH, et al. The adult human testis transcriptional cell atlas. Cell Res. 2018;28(12):1141-1157.

[9]

GuoJ, NieX, GieblerM, et al. The dynamic transcriptional cell atlas of testis development during human puberty. Cell Stem Cell. 2020;26:262-276.

[10]

VoigtAL, Dardari R, SuL, et al. Metabolic transitions define spermatogonial stem cell maturation. Hum Reprod. 2022;37(9):2095-2112.

[11]

ZhaoJ, LuP, WanC, et al. Cell-fate transition and determination analysis of mouse male germ cells throughout development. Nat Commun. 2021;12(1):6839.

[12]

HwangYS, SuzukiS, SeitaY, et al. Reconstitution of prospermatogonial specification in vitro from human induced pluripotent stem cells. Nat Commun. 2020;11(1):5656.

[13]

TangWW, Kobayashi T, IrieN, DietmannS, SuraniMA. Specification and epigenetic programming of the human germ line. Nat Rev Genet. 2016;17(10):585-600.

[14]

SaitouM, Miyauchi H. Gametogenesis from pluripotent stem cells. Cell Stem Cell. 2016;18(6):721-735.

[15]

SohniA, TanK, SongHW, et al. The neonatal and adult human testis defined at the single-cell level. Cell Rep. 2019;26(6):1501-1517.

[16]

TanK, Wilkinson MF. A single-cell view of spermatogonial stem cells. Curr Opin Cell Biol. 2020;67:71-78.

[17]

TanK, SongHW, WilkinsonMF. Single-cell RNAseq analysis of testicular germ and somatic cell development during the perinatal period. Development. 2020;147(3):dev183251.

[18]

ZhaoL, YaoC, XingX, et al. Single-cell analysis of developing and azoospermia human testicles reveals central role of Sertoli cells. Nat Commun. 2020;11(1):5683.

[19]

DobinA, DavisCA, SchlesingerF, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15-21.

[20]

HaoY, HaoS, Andersen-NissenE, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184(13):3573-3587 e3529.

[21]

JinS, Guerrero-Juarez CF, ZhangL, et al. Inference and analysis of cell-cell communication using CellChat. Nat Commun. 2021;12(1):1088.

[22]

BrowaeysR, Saelens W, SaeysY. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat Methods. 2019;17:159-162.

[23]

LiuX, SongJ, ZhangH, et al. Immune checkpoint HLA-E:CD94-NKG2A mediates evasion of circulating tumor cells from NK cell surveillance. Cancer Cell. 2023;41(2):272-287 e279.

[24]

LakeBB, MenonR, WinfreeS, et al. An atlas of healthy and injured cell states and niches in the human kidney. Nature. 2023;619(7970):585-594.

[25]

AibarS, Gonzalez-Blas CB, MoermanT, et al. SCENIC: single-cell regulatory network inference and clustering. Nat Methods. 2017;14(11):1083-1086.

[26]

JoungJ, MaS, TayT, et al. A transcription factor atlas of directed differentiation. Cell. 2023;186(1):209-229 e226.

[27]

HuJ, ZhangL, XiaH, et al. Tumor microenvironment remodeling after neoadjuvant immunotherapy in non-small cell lung cancer revealed by single-cell RNA sequencing. Genome Med. 2023;15(1):14.

[28]

MorabitoS, ReeseF, RahimzadehN, Miyoshi E, SwarupV. hdWGCNA identifies co-expression networks in high-dimensional transcriptomics data. Cell Rep Methods. 2023;3(6):100498.

[29]

McCarreyJR. Toward a more precise and informative nomenclature describing fetal and neonatal male germ cells in rodents. Biol Reprod. 2013;89(2):47.

[30]

YamajiM, SekiY, KurimotoK, et al. Critical function of Prdm14 for the establishment of the germ cell lineage in mice. Nat Genet. 2008;40(8):1016-1022.

[31]

SuzukiA, SagaY. Nanos2 suppresses meiosis and promotes male germ cell differentiation. Genes Dev. 2008;22(4):430-435.

[32]

Fend-GuellaDL, von Kopylow K, SpiessAN, et al. The DNA methylation profile of human spermatogonia at single-cell-and single-allele-resolution refutes its role in spermatogonial stem cell function and germ cell differentiation. Mol Hum Reprod. 2019;25(6):283-294.

[33]

MatsonCK, MurphyMW, GriswoldMD, Yoshida S, BardwellVJ, ZarkowerD. The mammalian doublesex homolog DMRT1 is a transcriptional gatekeeper that controls the mitosis versus meiosis decision in male germ cells. Dev Cell. 2010;19(4):612-624.

[34]

DingJ, Adiconis X, SimmonsSK, et al. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat Biotechnol. 2020;38(6):737-746.

[35]

WangX, HeY, ZhangQ, Ren X, ZhangZ. Direct comparative analyses of 10X genomics chromium and smart-seq2. Genomics Proteomics Bioinformatics. 2021;19(2):253-266.

[36]

PerchardeM, WongP, Ramalho-SantosM. Global Hypertranscription in the mouse embryonic germline. Cell Rep. 2017;19(10):1987-1996.

[37]

HayashiY, MoriM, IgarashiK, et al. Proteomic and metabolomic analyses uncover sex-specific regulatory pathways in mouse fetal germline differentiationdagger. Biol Reprod. 2020;103(4):717-735.

[38]

HayashiY, MatsuiY. Metabolic control of germline formation and differentiation in mammals. Sex Dev. 2022;16(5–6):388-403.

[39]

HayashiY, OtsukaK, EbinaM, et al. Distinct requirements for energy metabolism in mouse primordial germ cells and their reprogramming to embryonic germ cells. Proc Natl Acad Sci U S A. 2017;114(31):8289-8294.

[40]

WuYQ, ZhangCS, XiongJ, et al. Low glucose metabolite 3-phosphoglycerate switches PHGDH from serine synthesis to p53 activation to control cell fate. Cell Res. 2023;33(11):835-850.

[41]

RyazantsevS, Abuladze N, NewmanD, BondarG, KurtzI, PushkinA. Structural characterization of dimeric murine aminoacylase III. FEBS Lett. 2007;581(9):1898-1902.

[42]

HsiehJM, Tsirulnikov K, SawayaMR, et al. Structures of aminoacylase 3 in complex with acetylated substrates. Proc Natl Acad Sci U S A. 2010;107(42):17962-17967.

[43]

KulibinAY, Malolina EA. Formation of the rete testis during mouse embryonic development. Dev Dynam. 2020;249(12):1486-1499.

[44]

SvingenT, Koopman P. Building the mammalian testis: origins, differentiation, and assembly of the component cell populations. Genes Dev. 2013;27(22):2409-2426.

[45]

MauduitC, Hamamah S, BenahmedM. Stem cell factor/c-kit system in spermatogenesis. Hum Reprod Update. 1999;5(5):535-545.

[46]

MithraprabhuS, Loveland KL. Control of KIT signalling in male germ cells: what can we learn from other systems? Reproduction. 2009;138(5):743-757.

[47]

JiangEP, ZhangYQ. The role of transforming growth factor beta superfamily in male germ cell development. Zhonghua Nan Ke Xue. 2002;8(6):435-437.

[48]

MurtaD, Batista M, TrindadeA, et al. In vivo notch signaling blockade induces abnormal spermatogenesis in the mouse. PLoS One. 2014;9(11):e113365.

[49]

WuX, GaoS, WangL, et al. Role of laminin and collagen chains in human spermatogenesis—insights from studies in rodents and scRNA-Seq transcriptome profiling. Semin Cell Dev Biol. 2022;121:125-132.

[50]

de SantaBP, MoniotB, PoulatF, Berta P. Expression and subcellular localization of SF-1, SOX9, WT1, and AMH proteins during early human testicular development. Dev Dynam. 2000;217(3):293-298.

[51]

GaoY, WangZ, LongY, et al. Unveiling the roles of Sertoli cells lineage differentiation in reproductive development and disorders: a review. Front Endocrinol. 2024;15:1357594.

[52]

YefimovaMG, RavelC, RollandAD, Bourmeyster N, JegouB. MERTK-mediated LC3-associated phagocytosis (LAP) of apoptotic substrates in blood-separated tissues: retina, testis, ovarian follicles. Cells. 2021;10(6):1443.

[53]

RegevA, Goldman S, ShalevE. Semaphorin-4D (Sema-4D), the Plexin-B. ligand, is involved in mouse ovary follicular development. Reprod Biol Endocrinol. 2007;5:12.

[54]

BasuS, AryaSP, UsmaniA, et al. Defective Wnt3 expression by testicular Sertoli cells compromise male fertility. Cell Tissue Res. 2018;371(2):351-363.

[55]

FuY, WeiY, ZhouY, et al. Wnt5a regulates junctional function of Sertoli cells through PCP-mediated effects on mTORC1 and mTORC2. Endocrinology. 2021;162(10):bqab149.

[56]

WuW, HuY, ZhangQ, Xu Y, SuW. TNFalpha stimulates the proliferation of immature Sertoli cells by attenuating UPS-degradation of cyclin D1 and leads to the delay of BTB maturation in pubertal rats. Andrology. 2023;11(3):575-590.

[57]

Cartier-MichaudA, Bailly AL, BetziS, et al. Genetic, structural, and chemical insights into the dual function of GRASP55 in germ cell Golgi remodeling and JAM-C polarized localization during spermatogenesis. PLoS Genet. 2017;13(6):e1006803.

[58]

WuY, DuanP, WenY, et al. UHRF1 establishes crosstalk between somatic and germ cells in male reproduction. Cell Death Dis. 2022;13(4):377.

[59]

SzczepnyA, Hogarth CA, YoungJ, LovelandKL. Identification of hedgehog signaling outcomes in mouse testis development using a hanging drop-culture system. Biol Reprod. 2009;80(2):258-263.

[60]

ChungJW, PaskAJ, RenfreeMB. Seminiferous cord formation is regulated by hedgehog signaling in the marsupial. Biol Reprod. 2012;86(3):80.

[61]

GnessiL, EmidiA, ScarpaS, et al. Platelet-derived growth factor effects on purified testicular peritubular myoid cells: binding, cytosolic Ca2+ increase, mitogenic activity, and extracellular matrix production enhancement. Endocrinology. 1993;133(4):1880-1890.

[62]

JezekD, Hittmair A, RogatschH, KosM. Lamina propria of sex cords in human fetal testis: an immunohistological and stereological study. Anat Embryol. 1996;193(2):181-190.

[63]

ShenW, ParkBW, TomsD, Li J. Midkine promotes proliferation of primordial germ cells by inhibiting the expression of the deleted in azoospermia-like gene. Endocrinology. 2012;153(7):3482-3492.

[64]

HeY, ZhangB, HanD, et al. TAGLN2 promotes the proliferation, migration, invasion, and EMT of clear cell renal cell carcinoma through the PI3K/Akt signaling pathway. Biochem Genet. 2023;61(4):1265-1281.

[65]

KimHR, KwonMS, LeeS, et al. TAGLN2 polymerizes G-actin in a low ionic state but blocks Arp2/3-nucleated actin branching in physiological conditions. Sci Rep. 2018;8(1):5503.

[66]

NaBR, KimHR, PiragyteI, et al. TAGLN2 regulates T cell activation by stabilizing the actin cytoskeleton at the immunological synapse. J Cell Biol. 2015;209(1):143-162.

[67]

NaBR, JunCD. TAGLN2-mediated actin stabilization at the immunological synapse: implication for cytotoxic T cell control of target cells. BMB Rep. 2015;48(7):369-370.

[68]

JeongHS, BhinJ, Joon KimH, Hwang D, Ryul LeeD, KimKS. Transcriptional regulatory networks underlying the reprogramming of spermatogonial stem cells to multipotent stem cells. Exp Mol Med. 2017;49(4):e315.

[69]

GashawI, Grummer R, Klein-HitpassL, et al. Gene signatures of testicular seminoma with emphasis on expression of ets variant gene 4. Cellular and Molecular Life Sciences: CMLS. 2005;62(19–20):2359-2368.

[70]

AzarniadR, RaziM, HasanzadehS, Malekinejad H. Experimental diabetes negatively affects the spermatogonial stem cells’ self-renewal by suppressing GDNF network interactions. Andrologia. 2020;52(9):e13710.

[71]

LaHM, ChanAL, LegrandJMD, et al. GILZ-dependent modulation of mTORC1 regulates spermatogonial maintenance. Development. 2018;145(18):dev165324.

[72]

KaucherAV, OatleyMJ, OatleyJM. NEUROG3 is a critical downstream effector for STAT3-regulated differentiation of mammalian stem and progenitor spermatogonia. Biol Reprod. 2012;86(5):164.

[73]

IshiiK, Kanatsu-Shinohara M, ToyokuniS, ShinoharaT. FGF2 mediates mouse spermatogonial stem cell self-renewal via upregulation of Etv5 and Bcl6b through MAP2K1 activation. Development. 2012;139(10):1734-1743.

[74]

DannCT, Alvarado AL, MolyneuxLA, DenardBS, Garbers DL, PorteusMH. Spermatogonial stem cell self-renewal requires OCT4, a factor downregulated during retinoic acid-induced differentiation. Stem Cells. 2008;26(11):2928-2937.

[75]

ZhaoJY, FengKR, WangF, et al. A retrospective overview of PHGDH and its inhibitors for regulating cancer metabolism. Eur J Med Chem. 2021;217:113379.

[76]

ShunxiW, Xiaoxue Y, GuanbinS, LiY, JunyuJ, WanqianL. Serine metabolic reprogramming in tumorigenesis, tumor immunity, and clinical treatment. Adv Nutr. 2023;14(5):1050-1066.

[77]

YoshidaK, FuruyaS, OsukaS, et al. Targeted disruption of the mouse 3-phosphoglycerate dehydrogenase gene causes severe neurodevelopmental defects and results in embryonic lethality. J Biol Chem. 2004;279(5):3573-3577.

[78]

TsirulnikovK, DuarteS, RayA, et al. Aminoacylase 3 is a new potential marker and therapeutic target in hepatocellular carcinoma. J Cancer. 2018;9(1):1-12.

[79]

GallardoT, Shirley L, JohnGB, CastrillonDH. Generation of a germ cell-specific mouse transgenic Cre line, vasa-Cre. Genesis. 2007;45(6):413-417.

[80]

Sadate-NgatchouPI, Payne CJ, DearthAT, BraunRE. Cre recombinase activity specific to postnatal, premeiotic male germ cells in transgenic mice. Genesis. 2008;46(12):738-742.

[81]

SeisenbergerS, Andrews S, KruegerF, et al. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol Cell. 2012;48(6):849-862.

[82]

MolaroA, Falciatori I, HodgesE, et al. Two waves of de novo methylation during mouse germ cell development. Genes Dev. 2014;28(14):1544-1549.

[83]

YamanakaS, Nishihara H, TohH, et al. Broad heterochromatic domains open in Gonocyte development prior to De novo DNA methylation. Dev Cell. 2019;51(1):21-34 e25.

[84]

SachsM, Onodera C, BlaschkeK, EbataKT, SongJS, Ramalho-SantosM. Bivalent chromatin marks developmental regulatory genes in the mouse embryonic germline in vivo. Cell Rep. 2013;3(6):1777-1784.

[85]

HuangT-C, WangY-F. Vazquez-FerrerE, et al. Sex-specific chromatin remodelling safeguards transcription in germ cells. Nature. 2021;600:737-742.

[86]

ShiraneK, MiuraF, ItoT, Lorincz MC. NSD1-deposited H3K36me2 directs de novo methylation in the mouse male germline and counteracts Polycomb-associated silencing. Nat Genet. 2020;52(10):1088-1098.

[87]

SinghP, LiAX, TranDA, et al. De novo DNA methsylation in the male germ line occurs by default but is excluded at sites of H3K4 methylation. Cell Rep. 2013;4(1):205-219.

RIGHTS & PERMISSIONS

2024 The Author(s). Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.

AI Summary AI Mindmap
PDF

238

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/