Browse > Article

Temporal Expression of RNA Polymerase II in Porcine Oocytes and Embryos  

Oqani, Reza (Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University)
Lee, Min Gu (Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University)
Tao, Lin (Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University)
Jin, Dong Il (Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University)
Publication Information
Abstract
Embryonic genome activation (EGA) is the first major transition that occurs after fertilization, and entails a dramatic reprogramming of gene expression that is essential for continued development. Although it has been suggested that EGA in porcine embryos starts at the four-cell stage, recent evidence indicates that EGA may commence even earlier; however, the molecular details of EGA remain incompletely understood. The RNA polymerase II of eukaryotes transcribes mRNAs and most small nuclear RNAs. The largest subunit of RNA polymerase II can become phosphorylated in the C-terminal domain. The unphosphorylated form of the RNA polymerase II largest subunit C-terminal domain (IIa) plays a role in initiation of transcription, and the phosphorylated form (IIo) is required for transcriptional elongation and mRNA splicing. In the present study, we explored the nuclear translocation, nuclear localization, and phosphorylation dynamics of the RNA polymerase II C-terminal domain in immature pig oocytes, mature oocytes, two-, four-, and eight-cell embryos, and the morula and blastocyst. To this end, we used antibodies specific for the IIa and IIo forms of RNA polymerase II to stain the proteins. Unphosphorylated RNA polymerase II stained strongly in the nuclei of germinal vesicle oocytes, whereas the phosphorylated form of the enzyme was confined to the chromatin of prophase I oocytes. After fertilization, both unphosphorylated and phosphorylated RNA polymerase II began to accumulate in the nuclei of early stage one-cell embryos, and this pattern was maintained through to the blastocyst stage. The results suggest that both porcine oocytes and early embryos are transcriptionally competent, and that transcription of embryonic genes during the first three cell cycles parallels expression of phosphorylated RNA polymerase II.
Keywords
Porcine oocyte and embryo; Embryonic genome activation; RNA polymerase II; Phosphorylation; Transcription;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Forlani S, Bonnerot C, Capgras S, Nicolas JF (19998): Relief of a repressed gene expression state in the mouse 1-cell embryo requires DNA replication. Development 125:3153-3166.
2 Kanka J (2003): Gene expression and chromatin structure in the pre-implantation embryo. Theriogenology 59:3-19.   DOI   ScienceOn
3 Oqani RK, Kim HR, Zhang JY, Diao YF, Jin DI (2011): The CDK9/Cyclin T1 subunits of P-TEFb in mouse oocytes and preimplantation embryos: a possible ro le in embryonic genome activation. BMC Developmental Biology 11:33   DOI
4 Oqani RK, Zhang JY, Lee MG, Diao YF, Ijn DI (2012): Phosphorylation status of RNA polymerase II carboxyl- terminal domain in porcine oocytes and early embryos. Asian-Australasian J Anim Sci 25:789-793.   DOI   ScienceOn
5 Pirngruber J, Shchebet A, Johnsen SA (2009): Insights into the function of the human P-TEFb component CDK9 in the regulation of chromatin modifications and co-transcriptional mRNA processing. Cell Cycle 8:3636-3642.   DOI   ScienceOn
6 Price DH (2000): P-TEFb, a cyclin-dependent kinase controlling elongation by RNA polymerase II. Mol Cell Biol 20:2629-2634.   DOI   ScienceOn
7 Bellier S, Chastant S, Adenot P, Vincent M, Renard JP, Bensaude O (1997): Nuclear translocation and carboxyl- terminal domain phosphorylation of RNA polymerase II delineate the two phases of zygotic gene activation in mammalian embryos. EMBO J 16:6250-6262.   DOI   ScienceOn
8 Bogolyubova IO (2011): Transcriptional activity of nuclei in 2-cell blocked mouse embryos. Tissue Cell 43: 262-265.   DOI   ScienceOn
9 Bouniol C, Nguyen E, Debey P (1995): Endogenous transcription occurs at the 1-cell stage in the mouse embryo. Exp Cell Res 218:57-62.   DOI   ScienceOn
10 Bres V, Yoh SM, Jones KA (2008): The multi-tasking P-TEFb complex. Curr Opin Cell Biol 20:334-340.   DOI   ScienceOn
11 Dey A, Nishiyama A, Karpova T, McNally J, Ozato K (2009): Brd4 marks select genes on mitotic chromatin and directs postmitotic transcription. Mol Biol Cell 20:4899-4909.   DOI
12 Sims RJ 3rd, Mandal SS, Reinberg D (2004): Recent highlights of RNA-polymerase-II-mediated transcription. Curr Opin Cell Biol 16:263-271.   DOI   ScienceOn
13 Reese JC (2003): Basal transcription factors. Curr Opin Genet Dev 13:114-118.   DOI   ScienceOn
14 Schultz RM (2002): The molecular foundations of the maternal to zygotic transition in the preimplantation embryo. Hum Reprod Update 8:323-331.   DOI   ScienceOn
15 Sikorski TW, Buratowski S (2009): The basal initiation machinery: beyond the general transcription factors. Curr Opin Cell Biol 21:344-351.   DOI   ScienceOn
16 Tan JH, Wang HL, Sun XS, Liu Y, Sui HS, Zhang J (2009): Chromatin configurations in the germinal vesicle of mammalian oocytes. Mol Hum Reprod 15: 1-9.   DOI   ScienceOn
17 Walser CD, Lipshitz HD (2011): Transcript clearance during the maternal-to-zygotic transition. Curr Opin Genet Develop 21:431-443.   DOI   ScienceOn
18 Wrenzycki C, Herrmann D, Niemann H (2007): Messenger RNA in oocytes and embryos in relation to embryo viability. Theriogenology 68: 77-83   DOI
19 Xie SQ, Martin S, Guillot PV, Bentley DL, Pombo A (2006): Splicing speckles are not reservoirs of RNA polymerase II, but contain an inactive form, phosphorylated on serine2 residues of the C-terminal domain. Mol Biol Cell 17:1723-1733.   DOI   ScienceOn
20 Yang Z, He N, Zhou Q (2008): Brd4 recruits PTEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression. Mol Cell Biol 28:967-976.   DOI   ScienceOn