Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-05-21T10:26:59.944Z Has data issue: false hasContentIssue false

Pum3 is dispensable for mouse oocyte maturation and embryo development in vitro

Published online by Cambridge University Press:  17 May 2023

TingTing Zhao
Affiliation:
State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
Wei Huang*
Affiliation:
Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, 570102, China
Kaibo Lin*
Affiliation:
Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200021, China
*
Corresponding author: Kaibo Lin. Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200021, China. Email: linkb@126.com. Wei Huang. Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, 570102, China. Email: huang1213@hainmc.edu.cn
Corresponding author: Kaibo Lin. Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200021, China. Email: linkb@126.com. Wei Huang. Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, 570102, China. Email: huang1213@hainmc.edu.cn

Summary

Pumilio3 (Pum3), an evolutionarily distant homologue of the classical RNA-binding protein PUF (PUMILIO and FBF) family member, is also involved in the process of RNA metabolism through post-transcriptional regulation. However, the functions of Pum3 in mouse oocyte maturation and preimplantation embryonic development have not been elucidated. By comparing RNA levels in different tissues, we found that Pum3 was widely expressed in multiple tissues, but moderately predominant in the ovary. Histochemical staining suggested that the PUM3 protein exhibits positive signals in oocytes, granulosa cells and theca cells of different follicle stages. Oocyte immunofluorescence results showed a slightly higher level of PUM3 protein in metaphase II compared with the germinal vesicle (GV) stage. After knockdown of Pum3 in GV oocytes using siRNA injection (siPUM3), no obvious defect was observed in the processes of GV breakdown and polar body extrusion during in vitro maturation (IVM) for the siPum3 oocytes. Compared with the control group, the siPUM3 group displayed no significant abnormality in the cleavage and blastocyst formation rate of these fertilized oocytes. Therefore, we can conclude that depletion of Pum3 does not affect mouse oocyte maturation and early embryonic development in vitro.

Type
Research Article
Copyright
© The Author(s), 2023. 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

Blackinton, J. G. and Keene, J. D. (2014). Post-transcriptional RNA regulons affecting cell cycle and proliferation. Seminars in Cell and Developmental Biology, 34, 4454. doi: 10.1016/j.semcdb.2014.05.014 CrossRefGoogle ScholarPubMed
Chen, G., Li, W., Zhang, Q. S., Regulski, M., Sinha, N., Barditch, J., Tully, T., Krainer, A. R., Zhang, M. Q. and Dubnau, J. (2008). Identification of synaptic targets of Drosophila pumilio. PLOS Computational Biology, 4(2), e1000026. doi: 10.1371/journal.pcbi.1000026 CrossRefGoogle ScholarPubMed
Chen, D., Zheng, W., Lin, A., Uyhazi, K., Zhao, H. and Lin, H. (2012). Pumilio 1 suppresses multiple activators of p53 to safeguard spermatogenesis. Current Biology: CB, 22(5), 420425. doi: 10.1016/j.cub.2012.01.039 CrossRefGoogle ScholarPubMed
Cho, H. C., Huang, Y., Hung, J. T., Hung, T. H., Cheng, K. C., Liu, Y. H., Kuo, M. W., Wang, S. H., Yu, A. L. and Yu, J. (2022). Puf-A promotes cancer progression by interacting with nucleophosmin in nucleolus. Oncogene, 41(8), 11551165. doi: 10.1038/s41388-021-02138-0 CrossRefGoogle ScholarPubMed
Edwards, T. A., Pyle, S. E., Wharton, R. P. and Aggarwal, A. K. (2001). Structure of Pumilio reveals similarity between RNA and peptide binding motifs. Cell, 105(2), 281289. doi: 10.1016/s0092-8674(01)00318-x CrossRefGoogle ScholarPubMed
Elguindy, M. M. and Mendell, J. T. (2021). NORAD-induced Pumilio phase separation is required for genome stability. Nature, 595(7866), 303308. doi: 10.1038/s41586-021-03633-w CrossRefGoogle ScholarPubMed
Fan, C. C., Lee, L. Y., Yu, M. Y., Tzen, C. Y., Chou, C. and Chang, M. S. (2013). Upregulated hPuf-A promotes breast cancer tumorigenesis. Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine, 34(5), 25572564. doi: 10.1007/s13277-013-0801-4 CrossRefGoogle ScholarPubMed
Forbes, A. and Lehmann, R. (1998). Nanos and Pumilio have critical roles in the development and function of Drosophila germline stem cells. Development, 125(4), 679690. doi: 10.1242/dev.125.4.679 CrossRefGoogle ScholarPubMed
Gennarino, V. A., Singh, R. K., White, J. J., De Maio, A., Han, K., Kim, J. Y., Jafar-Nejad, P., di Ronza, A., Kang, H., Sayegh, L. S., Cooper, T. A., Orr, H. T., Sillitoe, R. V. and Zoghbi, H. Y. (2015). Pumilio1 haploinsufficiency leads to SCA1-like neurodegeneration by increasing wild-type ataxin1 levels. Cell, 160(6), 10871098. doi: 10.1016/j.cell.2015.02.012 CrossRefGoogle ScholarPubMed
Goldstrohm, A. C., Hall, T. M. T. and McKenney, K. M. (2018). Post-transcriptional regulatory functions of mammalian pumilio proteins. Trends in Genetics: TIG, 34(12), 972990. doi: 10.1016/j.tig.2018.09.006 CrossRefGoogle ScholarPubMed
Jenkins, H. T., Baker-Wilding, R. and Edwards, T. A. (2009). Structure and RNA binding of the mouse Pumilio-2 PUF domain. Journal of Structural Biology, 167(3), 271276. doi: 10.1016/j.jsb.2009.06.007 CrossRefGoogle ScholarPubMed
Joshna, C. R., Saha, P., Atugala, D., Chua, G. and Muench, D. G. (2020). Plant PUF RNA-binding proteins: A wealth of diversity for post-transcriptional gene regulation. Plant Science: An International Journal of Experimental Plant Biology, 297, 110505. doi: 10.1016/j.plantsci.2020.110505 CrossRefGoogle ScholarPubMed
Ko, C. F., Chang, Y. C., Cho, H. C. and Yu, J. (2022). The Puf-A protein is required for primordial germ cell development. Cells, 11(9), 1476. doi: 10.3390/cells11091476 CrossRefGoogle ScholarPubMed
Kraemer, B., Crittenden, S., Gallegos, M., Moulder, G., Barstead, R., Kimble, J. and Wickens, M. (1999). NANOS-3 and FBF proteins physically interact to control the sperm-oocyte switch in Caenorhabditis elegans . Current Biology: CB, 9(18), 10091018. doi: 10.1016/s0960-9822(99)80449-7 CrossRefGoogle ScholarPubMed
Kresoja-Rakic, J. and Santoro, R. (2019). Nucleolus and rRNA gene chromatin in early embryo development. Trends in Genetics: TIG, 35(11), 868879. doi: 10.1016/j.tig.2019.06.005 CrossRefGoogle ScholarPubMed
Kuo, M. W., Wang, S. H., Chang, J. C., Chang, C. H., Huang, L. J., Lin, H. H., Yu, A. L., Li, W. H. and Yu, J. (2009). A novel puf-A gene predicted from evolutionary analysis is involved in the development of eyes and primordial germ-cells. PLOS ONE, 4(3), e4980. doi: 10.1371/journal.pone.0004980 CrossRefGoogle ScholarPubMed
Li, X., Zhu, M., Zang, M., Cao, D., Xie, Z., Liang, H., Bian, Z., Zhao, T., Hu, Z. and Xu, E. Y. (2022). PUMILIO-mediated translational control of somatic cell cycle program promotes folliculogenesis and contributes to ovarian cancer progression. Cellular and Molecular Life Sciences: CMLS, 79(5), 279. doi: 10.1007/s00018-022-04254-w CrossRefGoogle ScholarPubMed
Liang, X., Hart, K. J., Dong, G., Siddiqui, F. A., Sebastian, A., Li, X., Albert, I., Miao, J., Lindner, S. E. and Cui, L. (2018). Puf3 participates in ribosomal biogenesis in malaria parasites. Journal of Cell Science, 131(6). doi: 10.1242/jcs.212597 Google ScholarPubMed
Lin, H. and Spradling, A. C. (1997). A novel group of pumilio mutations affects the asymmetric division of germline stem cells in the Drosophila ovary. Development, 124(12), 24632476. doi: 10.1242/dev.124.12.2463 CrossRefGoogle ScholarPubMed
Lin, K., Zhang, S., Shi, Q., Zhu, M., Gao, L., Xia, W., Geng, B., Zheng, Z. and Xu, E. Y. (2018). Essential requirement of mammalian Pumilio family in embryonic development. Molecular Biology of the Cell, 29(24), 29222932. doi: 10.1091/mbc.E18-06-0369 CrossRefGoogle ScholarPubMed
Lin, K., Qiang, W., Zhu, M., Ding, Y., Shi, Q., Chen, X., Zsiros, E., Wang, K., Yang, X., Kurita, T. and Xu, E. Y. (2019). Mammalian Pum1 and Pum2 control body size via translational regulation of the cell cycle inhibitor Cdkn1b. Cell Reports, 26(9), 24342450.e6. doi: 10.1016/j.celrep.2019.01.111 CrossRefGoogle ScholarPubMed
Lin, H. W., Lee, J. Y., Chou, N. L., Shih, T. W. and Chang, M. S. (2021). Phosphorylation of PUF-A/PUM3 on Y259 modulates PUF-A stability and cell proliferation. PLOS ONE, 16(8), e0256282. doi: 10.1371/journal.pone.0256282 CrossRefGoogle ScholarPubMed
Lu, G., Dolgner, S. J. and Hall, T. M. (2009). Understanding and engineering RNA sequence specificity of PUF proteins. Current Opinion in Structural Biology, 19(1), 110115. doi: 10.1016/j.sbi.2008.12.009 CrossRefGoogle ScholarPubMed
Luo, W., Ke, H., Liu, R., Qin, Y., Mak, W., Ma, J., Zhao, S. and Chen, Z. J. (2018). Variation analysis of PUM1 gene in Chinese women with primary ovarian insufficiency. Journal of Assisted Reproduction and Genetics, 35(4), 727731. doi: 10.1007/s10815-017-1110-4 CrossRefGoogle ScholarPubMed
Mak, W., Fang, C., Holden, T., Dratver, M. B. and Lin, H. (2016). An important role of pumilio 1 in regulating the development of the mammalian female germline. Biology of Reproduction, 94(6), 134. doi: 10.1095/biolreprod.115.137497 CrossRefGoogle ScholarPubMed
Mak, W., Xia, J., Cheng, E. C., Lowther, K. and Lin, H. (2018). A role of Pumilio 1 in mammalian oocyte maturation and maternal phase of embryogenesis. Cell and Bioscience, 8, 54. doi: 10.1186/s13578-018-0251-1 CrossRefGoogle ScholarPubMed
Matoulkova, E., Michalova, E., Vojtesek, B. and Hrstka, R. (2012). The role of the 3′ untranslated region in post-transcriptional regulation of protein expression in mammalian cells. RNA Biology, 9(5), 563576. doi: 10.4161/rna.20231 CrossRefGoogle ScholarPubMed
Miles, W. O., Tschöp, K., Herr, A., Ji, J. Y. and Dyson, N. J. (2012). Pumilio facilitates miRNA regulation of the E2F3 oncogene. Genes and Development, 26(4), 356368. doi: 10.1101/gad.182568.111 CrossRefGoogle ScholarPubMed
Munschauer, M., Nguyen, C. T., Sirokman, K., Hartigan, C. R., Hogstrom, L., Engreitz, J. M., Ulirsch, J. C., Fulco, C. P., Subramanian, V., Chen, J., Schenone, M., Guttman, M., Carr, S. A. and Lander, E. S. (2018). The NORAD lncRNA assembles a topoisomerase complex critical for genome stability. Nature, 561(7721), 132136. doi: 10.1038/s41586-018-0453-z CrossRefGoogle ScholarPubMed
Najdrová, V., Stairs, C. W., Vinopalová, M., Voleman, L. and Doležal, P. (2020). The evolution of the puf superfamily of proteins across the tree of eukaryotes. BMC Biology, 18(1), 77. doi: 10.1186/s12915-020-00814-3 CrossRefGoogle ScholarPubMed
Naudin, C., Hattabi, A., Michelet, F., Miri-Nezhad, A., Benyoucef, A., Pflumio, F., Guillonneau, F., Fichelson, S., Vigon, I., Dusanter-Fourt, I. and Lauret, E. (2017). PUMILIO/FOXP1 signaling drives expansion of hematopoietic stem/progenitor and leukemia cells. Blood, 129(18), 24932506. doi: 10.1182/blood-2016-10-747436 CrossRefGoogle ScholarPubMed
Nüsslein-Volhard, R. L. C. (1987). Involvement of the pumilio gene in the transport of an abdominal signal in the Drosophila embryo. Nature.Google Scholar
Parisi, M. and Lin, H. (1999). The Drosophila pumilio gene encodes two functional protein isoforms that play multiple roles in germline development, gonadogenesis, oogenesis and embryogenesis. Genetics, 153(1), 235250. doi: 10.1093/genetics/153.1.235 CrossRefGoogle ScholarPubMed
Schultz, R. M., Stein, P. and Svoboda, P. (2018). The oocyte-to-embryo transition in mouse: Past, present, and future. Biology of Reproduction, 99(1), 160174. doi: 10.1093/biolre/ioy013 CrossRefGoogle ScholarPubMed
Silva, I. L. Z., Kohata, A. A. and Shigunov, P. (2022). Modulation and function of Pumilio proteins in cancer. Seminars in Cancer Biology, 86(3), 298309. doi: 10.1016/j.semcancer.2022.03.010 CrossRefGoogle ScholarPubMed
Son, S. H., Jang, S. Y. and Park, H. S. (2021). Functions of PUF family RNA-binding proteins in Aspergillus nidulans . Journal of Microbiology and Biotechnology, 31(5), 676685. doi: 10.4014/jmb.2101.01011 CrossRefGoogle ScholarPubMed
Takahiro, M., Kimura, Y., Nakano, T. and Yamaguchi, S. (2021). Ribosomal stress induces 2-cell embryo-like state transition of the mouse ESCs through p53 activation. Biochemical and Biophysical Research Communications, 579, 175180. doi: 10.1016/j.bbrc.2021.09.068 Google ScholarPubMed
Vessey, J. P., Schoderboeck, L., Gingl, E., Luzi, E., Riefler, J., Di Leva, F., Karra, D., Thomas, S., Kiebler, M. A. and Macchi, P. (2010). Mammalian Pumilio 2 regulates dendrite morphogenesis and synaptic function. Proceedings of the National Academy of Sciences of the United States of America, 107(7), 32223227. doi: 10.1073/pnas.0907128107 CrossRefGoogle ScholarPubMed
Wang, M., Ogé, L., Perez-Garcia, M. D., Hamama, L. and Sakr, S. (2018). The PUF protein family: Overview on PUF RNA targets, biological functions, and post transcriptional regulation. International Journal of Molecular Sciences, 19(2). doi: 10.3390/ijms19020410 Google ScholarPubMed
Wang, X., Zamore, P. D. and Hall, T. M. (2001). Crystal structure of a Pumilio homology domain. Molecular Cell, 7(4), 855865. doi: 10.1016/s1097-2765(01)00229-5 CrossRefGoogle ScholarPubMed
Xu, E. Y., Chang, R., Salmon, N. A. and Reijo Pera, R. A. (2007). A gene trap mutation of a murine homolog of the Drosophila stem cell factor Pumilio results in smaller testes but does not affect litter size or fertility. Molecular Reproduction and Development, 74(7), 912921. doi: 10.1002/mrd.20687 CrossRefGoogle ScholarPubMed
Yan, Z., Liang, H., Deng, L., Long, H., Chen, H., Chai, W., Suo, L., Xu, C., Kuang, Y., Wu, L., Lu, S. and Lyu, Q. (2015). Eight-shaped hatching increases the risk of inner cell mass splitting in extended mouse embryo culture. PLOS ONE, 10(12), e0145172. doi: 10.1371/journal.pone.0145172 CrossRefGoogle ScholarPubMed
Yoon, D. S., Lee, K. M., Choi, Y., Ko, E. A., Lee, N. H., Cho, S., Park, K. H., Lee, J. H., Kim, H. W. and Lee, J. W. (2022). TLR4 downregulation by the RNA-binding protein PUM1 alleviates cellular aging and osteoarthritis. Cell Death and Differentiation, 29(7), 13641378. doi: 10.1038/s41418-021-00925-6 CrossRefGoogle ScholarPubMed
Zhang, B., Gallegos, M., Puoti, A., Durkin, E., Fields, S., Kimble, J. and Wickens, M. P. (1997). A conserved RNA-binding protein that regulates sexual fates in the C. elegans hermaphrodite germ line. Nature, 390(6659), 477484. doi: 10.1038/37297 CrossRefGoogle Scholar
Zhang, M., Chen, D., Xia, J., Han, W., Cui, X., Neuenkirchen, N., Hermes, G., Sestan, N. and Lin, H. (2017). Post-transcriptional regulation of mouse neurogenesis by Pumilio proteins. Genes and Development, 31(13), 13541369. doi: 10.1101/gad.298752.117 CrossRefGoogle ScholarPubMed
Zhao, T., Xiao, T., Cao, D., Xia, W., Gao, L., Cheng, L., Zang, M., Li, X. and Xu, E. Y. (2022). Sertoli cell PUMILIO proteins modulate mouse testis size through translational control of cell cycle regulators. Biology of Reproduction, 107(1), 135147. doi: 10.1093/biolre/ioac118 CrossRefGoogle ScholarPubMed