Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

Single-cell RNA-sequencing analysis and characterisation of testicular cells in giant panda (Ailuropoda melanoleuca)

Yi Zheng https://orcid.org/0000-0003-0152-2671 A , Yuliang Liu B C , Rong Hou B C , Keyu Shi B , Yijiao Chen B , Tongying Feng B and Junhui An https://orcid.org/0000-0001-9985-4820 B C *
+ Author Affiliations
- Author Affiliations

A Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.

B Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan 610081, China.

C Sichuan Academy of Giant Panda, Chengdu, Sichuan 610000, China.

* Correspondence to: junhuian@panda.org.cn

Handling Editor: James Cummins

Reproduction, Fertility and Development 34(14) 933-943 https://doi.org/10.1071/RD22039
Published online: 12 August 2022

© 2022 The Author(s) (or their employer(s)). Published by CSIRO Publishing

Abstract

Context: The giant panda (Ailuropoda melanoleuca) is a rare and endangered species to be preserved in China. The giant panda has a low reproductive capacity, and due to the scarcity of samples, studies on testes from giant panda are very limited, with little knowledge about the process of spermatogenesis in this species.

Aims: To establish the gene expression profiles in cells from the testis of a giant panda.

Methods: The 10× Genomics single-cell RNA-sequencing platform was applied to cells from the testis of an adult giant panda.

Key results: We identified eight testicular cell types including six somatic and two germ cell types from our single-cell RNA-sequencing datasets. We also identified the differentially expressed genes (DEGs) in each cell type, and performed functional enrichment analysis for the identified testicular cell types. Furthermore, by immunohistochemistry we explored the protein localisation patterns of several marker genes in testes from giant panda.

Conclusions: Our study has for the first time established the gene expression profiles in cells from the testis of a giant panda.

Implications: Our data provide a reference catalogue for spermatogenesis and testicular cells in the giant panda, laying the foundation for future breeding and preservation of this endangered species.

Keywords: giant panda, molecular marker, scRNA-seq, spermatid, spermatocyte, spermatogenesis, spermatogonia, testis.


References

Becht, E, McInnes, L, Healy, J, Dutertre, C-A, Kwok, IWH, Ng, LG, Ginhoux, F, and Newell, EW (2018). Dimensionality reduction for visualizing single-cell data using UMAP. Nature Biotechnology 37, 38–44.
Dimensionality reduction for visualizing single-cell data using UMAP.Crossref | GoogleScholarGoogle Scholar |

Bhang, DH, Kim, B-J, Kim, BG, Schadler, K, Baek, K-H, Kim, YH, Hsiao, W, Ding, B-S, Rafii, S, Weiss, MJ, Chou, ST, Kolon, TF, Ginsberg, JP, Ryu, B-Y, and Ryeom, S (2018). Testicular endothelial cells are a critical population in the germline stem cell niche. Nature Communications 9, 4379.
Testicular endothelial cells are a critical population in the germline stem cell niche.Crossref | GoogleScholarGoogle Scholar |

Boehm EM, Gildenberg MS, Washington MT (2016) Chapter Seven - The many roles of PCNA in eukaryotic DNA replication. In ‘The enzymes. Vol. 39’. (Eds LS Kaguni, MT Oliveira) pp. 231–254. (Elsevier Press)

Cardoso-Moreira, M, Halbert, J, Valloton, D, Velten, B, Chen, C, Shao, Y, Liechti, A, Ascenção, K, Rummel, C, Ovchinnikova, S, Mazin, PV, Xenarios, I, Harshman, K, Mort, M, Cooper, DN, Sandi, C, Soares, MJ, Ferreira, PG, Afonso, S, Carneiro, M, Turner, JMA, VandeBerg, JL, Fallahshahroudi, A, Jensen, P, Behr, R, Lisgo, S, Lindsay, S, Khaitovich, P, Huber, W, Baker, J, Anders, S, Zhang, YE, and Kaessmann, H (2019). Gene expression across mammalian organ development. Nature 571, 505–509.
Gene expression across mammalian organ development.Crossref | GoogleScholarGoogle Scholar |

Chen, D-Y, Wen, D-C, Zhang, Y-P, Sun, Q-Y, Han, Z-M, Liu, Z-H, Shi, P, Li, J-S, Xiangyu, J-G, Lian, L, Kou, Z-H, Wu, Y-Q, Chen, Y-C, Wang, P-Y, and Zhang, H-M (2002). Interspecies implantation and mitochondria fate of panda-rabbit cloned embryos. Biology of Reproduction 67, 637–642.
Interspecies implantation and mitochondria fate of panda-rabbit cloned embryos.Crossref | GoogleScholarGoogle Scholar |

Chen, Q, Deng, T, and Han, D (2016). Testicular immunoregulation and spermatogenesis. Seminars in Cell & Developmental Biology 59, 157–165.
Testicular immunoregulation and spermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Chen, X, Che, D, Zhang, P, Li, X, Yuan, Q, Liu, T, Guo, J, Feng, T, Wu, L, Liao, M, He, Z, and Zeng, W (2017). Profiling of miRNAs in porcine germ cells during spermatogenesis. Reproduction 154, 789–798.
Profiling of miRNAs in porcine germ cells during spermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Choi, YH, and Kim, JK (2019). Dissecting cellular heterogeneity using single-cell RNA sequencing. Molecules and Cells 42, 189–199.
Dissecting cellular heterogeneity using single-cell RNA sequencing.Crossref | GoogleScholarGoogle Scholar |

Da Ros, M, Lehtiniemi, T, Olotu, O, Meikar, O, and Kotaja, N (2019). Enrichment of pachytene spermatocytes and spermatids from mouse testes using standard laboratory equipment. Journal of Visualized Experiments , e60271.
Enrichment of pachytene spermatocytes and spermatids from mouse testes using standard laboratory equipment.Crossref | GoogleScholarGoogle Scholar |

Di Persio, S, Saracino, R, Fera, S, Muciaccia, B, Esposito, V, Boitani, C, Berloco, BP, Nudo, F, Spadetta, G, Stefanini, M, de Rooij, DG, and Vicini, E (2017). Spermatogonial kinetics in humans. Development 144, 3430–3439.
Spermatogonial kinetics in humans.Crossref | GoogleScholarGoogle Scholar |

Fayomi, AP, and Orwig, KE (2018). Spermatogonial stem cells and spermatogenesis in mice, monkeys and men. Stem Cell Research 29, 207–214.
Spermatogonial stem cells and spermatogenesis in mice, monkeys and men.Crossref | GoogleScholarGoogle Scholar |

França, LR, Hess, RA, Dufour, JM, Hofmann, MC, and Griswold, MD (2016). The Sertoli cell: one hundred fifty years of beauty and plasticity. Andrology 4, 189–212.
The Sertoli cell: one hundred fifty years of beauty and plasticity.Crossref | GoogleScholarGoogle Scholar |

Glazkov, MV, and Mednikov, BM (1981). DNA synthesis in the process of rat spermatogenesis. Ontogenez 12, 470–476.

Green, CD, Ma, Q, Manske, GL, Shami, AN, Zheng, X, Marini, S, Moritz, L, Sultan, C, Gurczynski, SJ, Moore, BB, Tallquist, MD, Li, JZ, and Hammoud, SS (2018). A comprehensive roadmap of murine spermatogenesis defined by single-cell RNA-seq. Developmental Cell 46, 651–667.e10.
A comprehensive roadmap of murine spermatogenesis defined by single-cell RNA-seq.Crossref | GoogleScholarGoogle Scholar |

Guo, J, Grow, EJ, Yi, C, Mlcochova, H, Maher, GJ, Lindskog, C, Murphy, PJ, Wike, CL, Carrell, DT, Goriely, A, Hotaling, JM, and Cairns, BR (2017). Chromatin and single-cell RNA-seq profiling reveal dynamic signaling and metabolic transitions during human spermatogonial stem cell development. Cell Stem Cell 21, 533–546.e6.
Chromatin and single-cell RNA-seq profiling reveal dynamic signaling and metabolic transitions during human spermatogonial stem cell development.Crossref | GoogleScholarGoogle Scholar |

Guo, J, Grow, EJ, Mlcochova, H, Maher, GJ, Lindskog, C, Nie, X, Guo, Y, Takei, Y, Yun, J, Cai, L, Kim, R, Carrell, DT, Goriely, A, Hotaling, JM, and Cairns, BR (2018). The adult human testis transcriptional cell atlas. Cell Research 28, 1141–1157.
The adult human testis transcriptional cell atlas.Crossref | GoogleScholarGoogle Scholar |

Guo, J, Nie, X, Giebler, M, Mlcochova, H, Wang, Y, Grow, EJ, DonorConnect Kim, R, Tharmalingam, M, Matilionyte, G, Lindskog, C, Carrell, DT, Mitchell, RT, Goriely, A, Hotaling, JM, and Cairns, BR (2020). The dynamic transcriptional cell atlas of testis development during human puberty. Cell Stem Cell 26, 262–276.e4.
The dynamic transcriptional cell atlas of testis development during human puberty.Crossref | GoogleScholarGoogle Scholar |

Guo, J, Sosa, E, Chitiashvili, T, Nie, X, Rojas, EJ, Oliver, E, DonorConnect Plath, K, Hotaling, JM, Stukenborg, J-B, Clark, AT, and Cairns, BR (2021). Single-cell analysis of the developing human testis reveals somatic niche cell specification and fetal germline stem cell establishment. Cell Stem Cell 28, 764–778.e4.
Single-cell analysis of the developing human testis reveals somatic niche cell specification and fetal germline stem cell establishment.Crossref | GoogleScholarGoogle Scholar |

Hermann, BP, Cheng, K, Singh, A, Roa-De La Cruz, L, Mutoji, KN, Chen, I-C, Gildersleeve, H, Lehle, JD, Mayo, M, Westernströer, B, Law, NC, Oatley, MJ, Velte, EK, Niedenberger, BA, Fritze, D, Silber, S, Geyer, CB, Oatley, JM, and McCarrey, JR (2018). The mammalian spermatogenesis single-cell transcriptome, from spermatogonial stem cells to spermatids. Cell Reports 25, 1650–1667.e8.
The mammalian spermatogenesis single-cell transcriptome, from spermatogonial stem cells to spermatids.Crossref | GoogleScholarGoogle Scholar |

Jan, SZ, Hamer, G, Repping, S, de Rooij, DG, van Pelt, AMM, and Vormer, TL (2012). Molecular control of rodent spermatogenesis. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1822, 1838–1850.
Molecular control of rodent spermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Jan, SZ, Vormer, TL, Jongejan, A, Röling, MD, Silber, SJ, de Rooij, DG, Hamer, G, Repping, S, and van Pelt, AMM (2017). Unraveling transcriptome dynamics in human spermatogenesis. Development 144, 3659–3673.
Unraveling transcriptome dynamics in human spermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Kim, Y-H, Oh, M-G, Bhang, DH, Kim, B-J, Jung, S-E, Kim, S-M, Dohr, G, Kim, S-U, Ryeom, S, and Ryu, B-Y (2019). Testicular endothelial cells promote self-renewal of spermatogonial stem cells in rats. Biology of Reproduction 101, 360–367.
Testicular endothelial cells promote self-renewal of spermatogonial stem cells in rats.Crossref | GoogleScholarGoogle Scholar |

Lau, X, Munusamy, P, Ng, MJ, and Sangrithi, M (2020). Single-cell RNA sequencing of the cynomolgus macaque testis reveals conserved transcriptional profiles during mammalian spermatogenesis. Developmental Cell 54, 548–566.e7.
Single-cell RNA sequencing of the cynomolgus macaque testis reveals conserved transcriptional profiles during mammalian spermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Li, R, Fan, W, Tian, G, Zhu, H, He, L, Cai, J, Huang, Q, Cai, Q, Li, B, Bai, Y, Zhang, Z, Zhang, Y, Wang, W, Li, J, Wei, F, Li, H, Jian, M, Li, J, Zhang, Z, Nielsen, R, Li, D, Gu, W, Yang, Z, Xuan, Z, Ryder, OA, Leung, FC-C, Zhou, Y, Cao, J, Sun, X, Fu, Y, Fang, X, Guo, X, Wang, B, Hou, R, Shen, F, Mu, B, Ni, P, Lin, R, Qian, W, Wang, G, Yu, C, Nie, W, Wang, J, Wu, Z, Liang, H, Min, J, Wu, Q, Cheng, S, Ruan, J, Wang, M, Shi, Z, Wen, M, Liu, B, Ren, X, Zheng, H, Dong, D, Cook, K, Shan, G, Zhang, H, Kosiol, C, Xie, X, Lu, Z, Zheng, H, Li, Y, Steiner, CC, Lam, TT-Y, Lin, S, Zhang, Q, Li, G, Tian, J, Gong, T, Liu, H, Zhang, D, Fang, L, Ye, C, Zhang, J, Hu, W, Xu, A, Ren, Y, Zhang, G, Bruford, MW, Li, Q, Ma, L, Guo, Y, An, N, Hu, Y, Zheng, Y, Shi, Y, Li, Z, Liu, Q, Chen, Y, Zhao, J, Qu, N, Zhao, S, Tian, F, Wang, X, Wang, H, Xu, L, Liu, X, Vinar, T, Wang, Y, Lam, T-W, Yiu, S-M, Liu, S, Zhang, H, Li, D, Huang, Y, Wang, X, Yang, G, Jiang, Z, Wang, J, Qin, N, Li, L, Li, J, Bolund, L, Kristiansen, K, Wong, GK-S, Olson, M, Zhang, X, Li, S, Yang, H, Wang, J, and Wang, J (2010). The sequence and de novo assembly of the giant panda genome. Nature 463, 311–317.
The sequence and de novo assembly of the giant panda genome.Crossref | GoogleScholarGoogle Scholar |

Li, H, Huang, Q, Liu, Y, and Garmire, LX (2020a). Single cell transcriptome research in human placenta. Reproduction 160, R155–R167.
Single cell transcriptome research in human placenta.Crossref | GoogleScholarGoogle Scholar |

Li, L, Yang, R, Yin, C, and Kee, K (2020b). Studying human reproductive biology through single-cell analysis and in vitro differentiation of stem cells into germ cell-like cells. Human Reproduction Update 26, 670–688.
Studying human reproductive biology through single-cell analysis and in vitro differentiation of stem cells into germ cell-like cells.Crossref | GoogleScholarGoogle Scholar |

Liao, J, Ng, SH, Luk, AC, Suen, HC, Qian, Y, Lee, AWT, Tu, J, Fung, JCL, Tang, NLS, Feng, B, Chan, WY, Fouchet, P, Hobbs, RM, and Lee, TL (2019). Revealing cellular and molecular transitions in neonatal germ cell differentiation using single cell RNA sequencing. Development 146, dev174953.
Revealing cellular and molecular transitions in neonatal germ cell differentiation using single cell RNA sequencing.Crossref | GoogleScholarGoogle Scholar |

Liu, Y, Niu, M, Yao, C, Hai, Y, Yuan, Q, Liu, Y, Guo, Y, Li, Z, and He, Z (2015). Fractionation of human spermatogenic cells using STA-PUT gravity sedimentation and their miRNA profiling. Scientific Reports 5, 8084.
Fractionation of human spermatogenic cells using STA-PUT gravity sedimentation and their miRNA profiling.Crossref | GoogleScholarGoogle Scholar |

Luo, J, Megee, S, Rathi, R, and Dobrinski, I (2006). Protein gene product 9.5 is a spermatogonia-specific marker in the pig testis: application to enrichment and culture of porcine spermatogonia. Molecular Reproduction and Development 73, 1531–1540.
Protein gene product 9.5 is a spermatogonia-specific marker in the pig testis: application to enrichment and culture of porcine spermatogonia.Crossref | GoogleScholarGoogle Scholar |

Luo, J, Megee, S, and Dobrinski, I (2009). Asymmetric distribution of UCH-L1 in spermatogonia is associated with maintenance and differentiation of spermatogonial stem cells. Journal of Cellular Physiology 220, 460–468.
Asymmetric distribution of UCH-L1 in spermatogonia is associated with maintenance and differentiation of spermatogonial stem cells.Crossref | GoogleScholarGoogle Scholar |

Maekawa, M, Kamimura, K, and Nagano, T (1996). Peritubular myoid cells in the testis: their structure and function. Archives of Histology and Cytology 59, 1–13.
Peritubular myoid cells in the testis: their structure and function.Crossref | GoogleScholarGoogle Scholar |

McCarthy, DJ, Chen, Y, and Smyth, GK (2012). Differential expression analysis of multifactor RNA-seq experiments with respect to biological variation. Nucleic Acids Research 40, 4288–4297.
Differential expression analysis of multifactor RNA-seq experiments with respect to biological variation.Crossref | GoogleScholarGoogle Scholar |

Melé, M, Ferreira, PG, Reverter, F, DeLuca, DS, Monlong, J, Sammeth, M, Young, TR, Goldmann, JM, Pervouchine, DD, Sullivan, TJ, Johnson, R, Segrè, AV, Djebali, S, Niarchou, A, Consortium The GTEx Wright, FA, Lappalainen, T, Calvo, M, Getz, G, Dermitzakis, ET, Ardlie, KG, and Guigó, R (2015). The human transcriptome across tissues and individuals. Science 348, 660–665.
The human transcriptome across tissues and individuals.Crossref | GoogleScholarGoogle Scholar |

Morrison, SJ, and Spradling, AC (2008). Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell 132, 598–611.
Stem cells and niches: mechanisms that promote stem cell maintenance throughout life.Crossref | GoogleScholarGoogle Scholar |

Nagano, R, Tabata, S, Nakanishi, Y, Ohsako, S, Kurohmaru, M, and Hayashi, Y (2000). Reproliferation and relocation of mouse male germ cells (gonocytes) during prespermatogenesis. Anatomical Record 258, 210–220.
Reproliferation and relocation of mouse male germ cells (gonocytes) during prespermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Niedenberger, BA, and Geyer, CB (2018). Advanced immunostaining approaches to study early male germ cell development. Stem Cell Research 27, 162–168.
Advanced immunostaining approaches to study early male germ cell development.Crossref | GoogleScholarGoogle Scholar |

Scadden, DT (2006). The stem-cell niche as an entity of action. Nature 441, 1075–1079.
The stem-cell niche as an entity of action.Crossref | GoogleScholarGoogle Scholar |

Schell, C, Albrecht, M, Spillner, S, Mayer, C, Kunz, L, Köhn, FM, Schwarzer, U, and Mayerhofer, A (2010). 15-deoxy-Δ12-14-prostaglandin-J2 induces hypertrophy and loss of contractility in human testicular peritubular cells: implications for human male fertility. Endocrinology 151, 1257–1268.
15-deoxy-Δ12-14-prostaglandin-J2 induces hypertrophy and loss of contractility in human testicular peritubular cells: implications for human male fertility.Crossref | GoogleScholarGoogle Scholar |

Shah, MA, Xu, C, Wu, S, Zhao, W, Luo, H, Yi, C, Liu, W, and Cai, X (2018). Isolation and characterization of spermatogenic cells from cattle, yak and cattleyak. Animal Reproduction Science 193, 182–190.
Isolation and characterization of spermatogenic cells from cattle, yak and cattleyak.Crossref | GoogleScholarGoogle Scholar |

Shami, AN, Zheng, X, Munyoki, SK, Ma, Q, Manske, GL, Green, CD, Sukhwani, M, Orwig, KE, Li, JZ, and Hammoud, SS (2020). Single-cell RNA sequencing of human, macaque, and mouse testes uncovers conserved and divergent features of mammalian spermatogenesis. Developmental Cell 54, 529–547.e12.
Single-cell RNA sequencing of human, macaque, and mouse testes uncovers conserved and divergent features of mammalian spermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Soumillon, M, Necsulea, A, Weier, M, Brawand, D, Zhang, X, Gu, H, Barthès, P, Kokkinaki, M, Nef, S, Gnirke, A, Dym, M, de Massy, B, Mikkelsen, TS, and Kaessmann, H (2013). Cellular source and mechanisms of high transcriptome complexity in the mammalian testis. Cell Reports 3, 2179–2190.
Cellular source and mechanisms of high transcriptome complexity in the mammalian testis.Crossref | GoogleScholarGoogle Scholar |

Stuart, T, Butler, A, Hoffman, P, Hafemeister, C, Papalexi, E, Mauck, WM, Hao, Y, Stoeckius, M, Smibert, P, and Satija, R (2019). Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21.
Comprehensive integration of single-cell data.Crossref | GoogleScholarGoogle Scholar |

Suzuki, S, Diaz, VD, and Hermann, BP (2019). What has single-cell RNA-seq taught us about mammalian spermatogenesis? Biology of Reproduction 101, 617–634.
What has single-cell RNA-seq taught us about mammalian spermatogenesis?Crossref | GoogleScholarGoogle Scholar |

Tan, K, and Wilkinson, MF (2019). Human spermatogonial stem cells scrutinized under the single-cell magnifying glass. Cell Stem Cell 24, 201–203.
Human spermatogonial stem cells scrutinized under the single-cell magnifying glass.Crossref | GoogleScholarGoogle Scholar |

Tan, K, and Wilkinson, MF (2020). A single-cell view of spermatogonial stem cells. Current Opinion in Cell Biology 67, 71–78.
A single-cell view of spermatogonial stem cells.Crossref | GoogleScholarGoogle Scholar |

Tan, K, Song, H-W, and Wilkinson, MF (2020). Single-cell RNAseq analysis of testicular germ and somatic cell development during the perinatal period. Development 147, dev183251.
Single-cell RNAseq analysis of testicular germ and somatic cell development during the perinatal period.Crossref | GoogleScholarGoogle Scholar |

Wang, H, Wen, L, Yuan, Q, Sun, M, Niu, M, and He, Z (2016). Establishment and applications of male germ cell and Sertoli cell lines. Reproduction 152, R31–R40.
Establishment and applications of male germ cell and Sertoli cell lines.Crossref | GoogleScholarGoogle Scholar |

Wang, G, Li, Y, Yang, Q, Xu, S, Ma, S, Yan, R, Zhang, R, Jia, G, Ai, D, and Yang, Q (2019). Gene expression dynamics during the gonocyte to spermatogonia transition and spermatogenesis in the domestic yak. Journal of Animal Science and Biotechnology 10, 64.
Gene expression dynamics during the gonocyte to spermatogonia transition and spermatogenesis in the domestic yak.Crossref | GoogleScholarGoogle Scholar |

Xiang, L, Yin, Y, Zheng, Y, Ma, Y, Li, Y, Zhao, Z, Guo, J, Ai, Z, Niu, Y, Duan, K, He, J, Ren, S, Wu, D, Bai, Y, Shang, Z, Dai, X, Ji, W, and Li, T (2020). A developmental landscape of 3D-cultured human pre-gastrulation embryos. Nature 577, 537–542.
A developmental landscape of 3D-cultured human pre-gastrulation embryos.Crossref | GoogleScholarGoogle Scholar |

Yan, L, Yang, M, Guo, H, Yang, L, Wu, J, Li, R, Liu, P, Lian, Y, Zheng, X, Yan, J, Huang, J, Li, M, Wu, X, Wen, L, Lao, K, Li, R, Qiao, J, and Tang, F (2013). Single-cell RNA-seq profiling of human preimplantation embryos and embryonic stem cells. Nature Structural & Molecular Biology 20, 1131–1139.
Single-cell RNA-seq profiling of human preimplantation embryos and embryonic stem cells.Crossref | GoogleScholarGoogle Scholar |

Yang, H, Wang, F, Li, F, Ren, C, Pang, J, Wan, Y, Wang, Z, Feng, X, and Zhang, Y (2018). Comprehensive analysis of long noncoding RNA and mRNA expression patterns in sheep testicular maturation. Biology of Reproduction 99, 650–661.
Comprehensive analysis of long noncoding RNA and mRNA expression patterns in sheep testicular maturation.Crossref | GoogleScholarGoogle Scholar |

Yang, H, Ma, J, Wan, Z, Wang, Q, Wang, Z, Zhao, J, Wang, F, and Zhang, Y (2021). Characterization of sheep spermatogenesis through single-cell RNA sequencing. The FASEB Journal 35, e21187.
Characterization of sheep spermatogenesis through single-cell RNA sequencing.Crossref | GoogleScholarGoogle Scholar |

Yu, X-W, Li, T-T, Du, X-M, Shen, Q-Y, Zhang, M-F, Wei, Y-D, Yang, D-H, Xu, W-J, Chen, W-B, Bai, C-L, Li, X-L, Li, G-P, Li, N, Peng, S, Liao, M-Z, and Hua, J-L (2021). Single-cell RNA sequencing reveals atlas of dairy goat testis cells. Zoological Research 42, 401–405.
Single-cell RNA sequencing reveals atlas of dairy goat testis cells.Crossref | GoogleScholarGoogle Scholar |

Zhang, H, Li, D, Wang, C, and Hull, V (2009). Delayed implantation in giant pandas: the first comprehensive empirical evidence. Reproduction 138, 979–986.
Delayed implantation in giant pandas: the first comprehensive empirical evidence.Crossref | GoogleScholarGoogle Scholar |

Zhang, Q, Wang, Q, Zhang, Y, Cheng, S, Hu, J, Ma, Y, and Zhao, X (2018). Comprehensive analysis of microRNA–messenger rna from white yak testis reveals the differentially expressed molecules involved in development and reproduction. International Journal of Molecular Sciences 19, 3083.
Comprehensive analysis of microRNA–messenger rna from white yak testis reveals the differentially expressed molecules involved in development and reproduction.Crossref | GoogleScholarGoogle Scholar |

Zhang, P, Li, F, Zhang, L, Lei, P, Zheng, Y, and Zeng, W (2020). Stage-specific embryonic antigen 4 is a membrane marker for enrichment of porcine spermatogonial stem cells. Andrology 8, 1923–1934.
Stage-specific embryonic antigen 4 is a membrane marker for enrichment of porcine spermatogonial stem cells.Crossref | GoogleScholarGoogle Scholar |

Zhang, L, Li, F, Lei, P, Guo, M, Liu, R, Wang, L, Yu, T, Lv, Y, Zhang, T, Zeng, W, Lu, H, and Zheng, Y (2021). Single-cell RNA-sequencing reveals the dynamic process and novel markers in porcine spermatogenesis. Journal of Animal Science and Biotechnology 12, 122.
Single-cell RNA-sequencing reveals the dynamic process and novel markers in porcine spermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Zhang, L, Guo, M, Liu, Z, Liu, R, Zheng, Y, Yu, T, Lv, Y, Lu, H, Zeng, W, Zhang, T, and Pan, C (2022a). Single-cell RNA-seq analysis of testicular somatic cell development in pigs. Journal of Genetics and Genomics , .
Single-cell RNA-seq analysis of testicular somatic cell development in pigs.Crossref | GoogleScholarGoogle Scholar |

Zhang, L-K, Ma, H-D, Guo, M, Wang, L, Zheng, Y, Wu, X-D, Li, T-J, Lu, H-Z, Zeng, W-X, and Zhang, T (2022b). Dynamic transcriptional atlas of male germ cells during porcine puberty. Zoological Research 43, 600–603.
Dynamic transcriptional atlas of male germ cells during porcine puberty.Crossref | GoogleScholarGoogle Scholar |

Zheng, Y, He, Y, An, J, Qin, J, Wang, Y, Zhang, Y, Tian, X, and Zeng, W (2014a). THY1 is a surface marker of porcine gonocytes. Reproduction, Fertility and Development 26, 533–539.
THY1 is a surface marker of porcine gonocytes.Crossref | GoogleScholarGoogle Scholar |

Zheng, Y, Zhang, Y, Qu, R, He, Y, Tian, X, and Zeng, W (2014b). Spermatogonial stem cells from domestic animals: progress and prospects. Reproduction 147, R65–R74.
Spermatogonial stem cells from domestic animals: progress and prospects.Crossref | GoogleScholarGoogle Scholar |

Zheng, Y, Feng, T, Zhang, P, Lei, P, Li, F, and Zeng, W (2020). Establishment of cell lines with porcine spermatogonial stem cell properties. Journal of Animal Science and Biotechnology 11, 33.
Establishment of cell lines with porcine spermatogonial stem cell properties.Crossref | GoogleScholarGoogle Scholar |

Zheng, Y, Gao, Q, Li, T, Liu, R, Cheng, Z, Guo, M, Xiao, J, Wu, D, and Zeng, W (2022a). Sertoli cell and spermatogonial development in pigs. Journal of Animal Science and Biotechnology 13, 45.
Sertoli cell and spermatogonial development in pigs.Crossref | GoogleScholarGoogle Scholar |

Zheng, Y, Zhang, L, Jin, L, Zhang, P, Li, F, Guo, M, Gao, Q, Zeng, Y, Li, M, and Zeng, W (2022b). Unraveling three-dimensional chromatin structural dynamics during spermatogonial differentiation. Journal of Biological Chemistry 298, 101559.
Unraveling three-dimensional chromatin structural dynamics during spermatogonial differentiation.Crossref | GoogleScholarGoogle Scholar |

Zhu, Z-J, Yang, S, and Li, Z (2015). Transcriptome research on spermatogenic molecular drive in mammals. Asian Journal of Andrology 17, 961–971.
Transcriptome research on spermatogenic molecular drive in mammals.Crossref | GoogleScholarGoogle Scholar |

Zirkin, BR, and Papadopoulos, V (2018). Leydig cells: formation, function, and regulation. Biology of Reproduction 99, 101–111.
Leydig cells: formation, function, and regulation.Crossref | GoogleScholarGoogle Scholar |