Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-23T10:58:14.920Z Has data issue: false hasContentIssue false

Sperm-borne small RNAs improve the developmental competence of pre-implantation cloned embryos in rabbit

Published online by Cambridge University Press:  09 March 2021

Hongyu Qin
Affiliation:
Precision Medicine Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, China
Pengxiang Qu*
Affiliation:
Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China
Huizhong Hu
Affiliation:
Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China
Wenbin Cao
Affiliation:
Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China
Hengchao Liu
Affiliation:
Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China
Yanru Zhang
Affiliation:
Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China
Jinpeng Zhao
Affiliation:
Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China
Fatima Nazira
Affiliation:
Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China
Enqi Liu*
Affiliation:
Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China
*
Author for correspondence: Pengxiang Qu and Enqi Liu. Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China. E-mail: qupengxiang@xjtu.edu.cn, or liuenqi@xjtu.edu.cn
Author for correspondence: Pengxiang Qu and Enqi Liu. Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an, 710061, Shaanxi, China. E-mail: qupengxiang@xjtu.edu.cn, or liuenqi@xjtu.edu.cn

Summary

The low efficiency of somatic cell nuclear transfer (SCNT) greatly limits its application. Compared with the fertilized embryo, cloned embryos display abnormal epigenetic modification and other inferior developmental properties. In this study, small RNAs were isolated, and miR-34c and miR-125b were quantified by real-time PCR; results showed that these micro-RNAs were highly expressed in sperm. The test sample was divided into three groups: one was the fertilized group, one was the SCNT control group (NT-C group), and the third group consisted of SCNT embryos injected with sperm-borne small RNA (NT-T group). The level of tri-methylation of lysine 9 on histone H3 (H3K9me3) at the 8-cell stage was determined by immunofluorescence staining, and the cleavage ratio, blastocyst ratio, apoptotic cell index of the blastocyst and total cell number of blastocysts in each group were analyzed. Results showed that the H3K9me3 level was significantly higher in the NT-C group than in the fertilized group and the NT-T group. The apoptosis index of blastocysts in the NT-C group was significantly higher than that in the fertilized group and the NT-T group. The total cell number of SCNT embryos was significantly lower than that of fertilized embryos, and injecting sperm-borne small RNAs could significantly increase the total cell number of SCNT blastocysts. Our study not only demonstrates that sperm-borne small RNAs have an important role in embryo development, but also provides a new strategy for improving the efficiency of SCNT in rabbit.

Type
Research Article
Copyright
© The Author(s), 2021. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Bromhall, JD (1975). Nuclear transplantation in the rabbit egg. Nature 258, 719–22.CrossRefGoogle ScholarPubMed
Castellini, C, Mattioli, S, Bosco, AD, Cotozzolo, E, Cartoni Mancinelli, A, Rende, M, Stabile, AM and Pistilli, A (2020). Nerve growth factor receptor role on rabbit sperm storage. Theriogenology 153, 5461.CrossRefGoogle ScholarPubMed
Chen, Q, Yan, M, Cao, Z, Li, X, Zhang, Y, Shi, J, Feng, G-H, Peng, H, Zhang, X, Zhang, Y, Qian, J, Duan, E, Zhai, Q and Zhou, Q (2016a). Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science 351, 397400.CrossRefGoogle ScholarPubMed
Chen, Q, Yan, W and Duan, E (2016b). Epigenetic inheritance of acquired traits through sperm RNAs and sperm RNA modifications. Nat Rev Genet 17, 733–43.CrossRefGoogle ScholarPubMed
Chesné, P, Adenot, PG, Viglietta, C, Baratte, M, Boulanger, L and Renard, J-P (2002). Cloned rabbits produced by nuclear transfer from adult somatic cells. Nat Biotechnol 20, 366–9.CrossRefGoogle ScholarPubMed
Fouladi-Nashta, AA, Alberio, R, Kafi, M, Nicholas, B, Campbell, KHS and Webb, R (2005). Differential staining combined with TUNEL labelling to detect apoptosis in preimplantation bovine embryos. Reprod Biomed Online 10, 497502.CrossRefGoogle ScholarPubMed
Gapp, K, Jawaid, A, Sarkies, P, Bohacek, J, Pelczar, P, Prados, J, Farinelli, L, Miska, E and Mansuy, IM (2014). Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nat Neurosci 17, 667–9.CrossRefGoogle ScholarPubMed
Gonzalez-Munoz, E and Cibelli, JB (2018). Somatic cell reprogramming informed by the oocyte. Stem Cells Dev 27, 871–87.CrossRefGoogle ScholarPubMed
Gouveia, C, Huyser, C, Egli, D and Pepper, MS (2020). Lessons learned from somatic cell nuclear transfer. Int J Mol Sci 21, 2314.CrossRefGoogle ScholarPubMed
Honda, A and Ogura, A (2017). Rabbit models for biomedical research revisited via genome editing approaches. J Reprod Dev 63, 435–8.CrossRefGoogle ScholarPubMed
Jennane, S, El Haddad, M, Mahtat, EM, Doghmi, K and Mikdame, M (2017). Successful treatment of donor cell derived myelodysplastic syndrome with 5-azacytidine. Ann Biol Clin (Paris) 75, 713–4.Google ScholarPubMed
Jia, L, Ding, B, Shen, C, Luo, S, Zhang, Y, Zhou, L, Ding, R, Qu, P and Liu, E (2019). Use of oocytes selected by brilliant cresyl blue staining enhances rabbit cloned embryo development in vitro . Zygote 27, 166–72.CrossRefGoogle ScholarPubMed
Liu, W-M, Pang, RT K, Chiu, PCN, Wong, BPC, Lao, K, Lee, K-F and Yeung, WSB (2012). Sperm-borne microRNA-34c is required for the first cleavage division in mouse. Proc Natl Acad Sci USA 109, 490–4.CrossRefGoogle Scholar
Liu, X, Wang, Y, Gao, Y, Su, J, Zhang, J, Xing, X, Zhou, C, Yao, K, An, Q and Zhang, Y (2018). H3K9 demethylase KDM4E is an epigenetic regulator for bovine embryonic development and a defective factor for nuclear reprogramming. Development 145, dev158261.CrossRefGoogle Scholar
Ma, X, Zhang, Y, Qiao, F, Wang, M, Zuo, Z, Zhang, Y and Wang, Y (2018). Comparison of RNA extraction and microRNA detection protocols for a small amount of germinal vesicle oocytes in bovine. Anim Reprod Sci 195, 112–20.CrossRefGoogle ScholarPubMed
Matoba, S and Zhang, Y (2018). Somatic cell nuclear transfer reprogramming: mechanisms and applications. Cell Stem Cell 23, 471–85.CrossRefGoogle ScholarPubMed
Miyamoto, K, Hoshino, Y, Minami, N, Yamada, M and Imai, H (2007). Effects of synchronization of donor cell cycle on embryonic development and DNA synthesis in porcine nuclear transfer embryos. J Reprod Dev 53, 237–46.CrossRefGoogle ScholarPubMed
Nicetto, D and Zaret, KS (2019). Role of H3K9me3 heterochromatin in cell identity establishment and maintenance. Curr Opin Genet Dev 55, 110.CrossRefGoogle ScholarPubMed
Parrington, J, Arnoult, C and Fissore, RA (2019). The eggstraordinary story of how life begins. Mol Reprod Dev 86, 419.CrossRefGoogle ScholarPubMed
Parthipan, S, Selvaraju, S, Somashekar, L, Kolte, AP, Arangasamy, A and Ravindra, JP (2015). Spermatozoa input concentrations and RNA isolation methods on RNA yield and quality in bull (Bos taurus). Anal Biochem 482, 32–9.CrossRefGoogle Scholar
Qu, P, Zuo, Z, Liu, Z, Niu, Z, Zhang, Y, Du, Y, Ma, X, Qiao, F, Wang, M, Zhang, Y, Qing, S and Wang, Y (2019). Sperm-borne small RNAs regulate α-tubulin acetylation and epigenetic modification of early bovine somatic cell nuclear transfer embryos. Mol Hum Reprod 25, 471–82.CrossRefGoogle ScholarPubMed
Qu, P, Wang, Y, Zhang, C and Liu, E (2020a). Insights into the roles of sperm in animal cloning. Stem Cell Res Ther 11, 65.CrossRefGoogle ScholarPubMed
Qu, P, Shen, C, Du, Y, Qin, H, Luo, S, Fu, S, Dong, Y, Guo, S, Hu, F, Xue, Y and Liu, E (2020b). Melatonin protects rabbit somatic cell nuclear transfer (SCNT) embryos from electrofusion damage. Sci Rep 10, 2186.CrossRefGoogle ScholarPubMed
Rodgers, AB, Morgan, CP, Leu, NA and Bale, TL (2015). Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress. Proc Natl Acad Sci USA 112, 13699–704.CrossRefGoogle ScholarPubMed
Schuster, A, Tang, C, Xie, Y, Ortogero, N, Yuan, S and Yan, W (2016). SpermBase: a database for sperm-borne RNA contents. Biol Reprod 95, 99.CrossRefGoogle ScholarPubMed
Selokar, NL, St John, L, Revay, T, King, WA, Singla, SK and Madan, P (2013). Effect of histone deacetylase inhibitor valproic acid treatment on donor cell growth characteristics, cell cycle arrest, apoptosis, and handmade cloned bovine embryo production efficiency. Cell Reprogram 15, 531–42.CrossRefGoogle ScholarPubMed
Skinner, MK, Ben Maamar, M, Sadler-Riggleman, I, Beck, D, Nilsson, E, McBirney, M, Klukovich, R, Xie, Y, Tang, C and Yan, W (2018). Alterations in sperm DNA methylation, non-coding RNA and histone retention associate with DDT-induced epigenetic transgenerational inheritance of disease. Epigenetics Chromatin 11, 8.CrossRefGoogle ScholarPubMed
Song, X, Liu, Z, He, H, Wang, J, Li, H, Li, J, Li, F, Jiang, Z and Huan, Y (2017). Dnmt1s in donor cells is a barrier to SCNT-mediated DNA methylation reprogramming in pigs. Oncotarget 8, 34980–91.CrossRefGoogle ScholarPubMed
Sun, Z, Zhang, W, Li, S, Xue, X, Niu, R, Shi, L, Li, B, Wang, X and Wang, J (2016). Altered miRNAs expression profiling in sperm of mice induced by fluoride. Chemosphere 155, 109–14.CrossRefGoogle ScholarPubMed
Tosti, E and Ménézo, Y (2016). Gamete activation: basic knowledge and clinical applications. Hum Reprod Update 22, 420–39.CrossRefGoogle ScholarPubMed
van der Merwe, J, van der Veeken, L, Ferraris, S, Gsell, W, Himmelreich, U, Toelen, J, Ourselin, S, Melbourne, A, Vercauteren, T and Deprest, J (2019). Publisher Correction: Early neuropathological and neurobehavioral consequences of preterm birth in a rabbit model. Sci Rep 9, 17005.CrossRefGoogle ScholarPubMed
Wilmut, I, Schnieke, AE, McWhir, J, Kind, AJ and Campbell, KHS (1997). Viable offspring derived from fetal and adult mammalian cells. Nature 385, 810–3.CrossRefGoogle ScholarPubMed
Zhang, J, Qu, P, Zhou, C, Liu, X, Ma, X, Wang, M, Wang, Y, Su, J, Liu, J and Zhang, Y (2017). MicroRNA-125b is a key epigenetic regulatory factor that promotes nuclear transfer reprogramming. J Biol Chem 292, 15916–26.CrossRefGoogle ScholarPubMed
Zhang, Y, Shi, J, Rassoulzadegan, M, Tuorto, F and Chen, Q (2019). Sperm RNA code programmes the metabolic health of offspring. Nat Rev Endocrinol 15, 489–98.CrossRefGoogle ScholarPubMed