Int J Stem Cells.  2014 Nov;7(2):162-166. 10.15283/ijsc.2014.7.2.162.

Temporal and Spatial Expression Patterns of miR-302 and miR-367 During Early Embryonic Chick Development

Affiliations
  • 1Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Korea. ks66kim@hanyang.ac.kr
  • 2Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Korea. hsjung@yuhs.ac
  • 3Department of Radiology, Seoul National University Hospital, Seoul, Korea.
  • 4Oral Biosciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR.

Abstract

The microRNAs (miRNAs) are small, non-coding RNAs that modulate protein expression by interfering with target mRNA translation or stability. miRNAs play crucial roles in various functions such as cellular, developmental, and physiological processes. The spatial expression patterns of miRNAs are very essential for identifying their functions. The expressions of miR-302 and miR-367 are critical in maintaining stemness of pluripotent stem cells, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) but their functions in early development are not fully elucidated. So, we used Locked Nucleic Acid (LNA) probes to perform in situ hybridization and confirmed the temporal and spatial distribution patterns during early chick development. As a result, we found that miR-302 and miR-367 were expressed in various tissues such as primitive steak, neural ectoderm, neural plate, neural fold, neural tube, notochord, and oral cavity. Specially, we confirmed that miR-302 and miR-367 were strongly expressed in neural folds in HH8 to HH10. miR-302 was expressed on dorsal part of the neural tube but miR-367 was expressed on lateral and ventral parts of the neural tube. And also we performed quantitative stem-loop real-time PCR to analyze global expression level of miR-302 and miR-367. miR-302 and miR-367 expression was sustained before Hamburger and Hamilton stage (HH) 14. Thus, the temporal and spatial expression patterns of miR-302 and miR-367 may provide us information of the role of these miRNAs on tissue formation during early chick development.

Keyword

Early chick development; miRNAs; LNA; In situ hybridization

MeSH Terms

Ectoderm
Embryonic Stem Cells
In Situ Hybridization
Induced Pluripotent Stem Cells
MicroRNAs
Mouth
Neural Crest
Neural Plate
Neural Tube
Notochord
Physiological Processes
Pluripotent Stem Cells
Protein Biosynthesis
Real-Time Polymerase Chain Reaction
RNA, Untranslated
MicroRNAs
RNA, Untranslated

Figure

  • Fig. 1. Expression patterns and level of miR-302 during early chick development. Embryos expressed miR-302 by HH stages 4–13 (A–F). Black lines indicate level of tranverse sections presented in right or left beside of each embryo. miR-302 was expressed in epiblast (A1, B1), primitive streak (A2, B2, C2, D arrowhead), neural plate (C1, D1, D2), neural fold (C1, D1, D2), prosencephalon (E1), oral cavity (E1), neural tube (E2), and notochord (F1). Expression level of miR-302 was much higher at HH5 compared with other stages Relative expression value of miR-302 was normalized to the level of 18s mRNA(G). Bars represent mean±SEM. Student’s t-test: *p<0.05 and ***p<0.001 vs. HH1. Abbreviations: epi, epiblast; hyp, hyperblast; fg, foregut; nc, notochord; np, neural plate; nf, neural fold; nt, neurlal tube; oc, oral cavity; ps, primitive streak; pros, prosencephalon; sm, somite; HH, Hamburger and Hamilton.

  • Fig. 2. Differential expression patterns and level of miR-367 in early chick embryos. miR-367 was expressed in epiblast (A1, B1), primitive streak (A2, B2, C2, D arrowhead), neural plate (C1, D1), neural fold (C1, D1), prosencephalon (E1), Oral cavity (E1), neural tube (E2), notochord (D1, F1). Higher expression of miR-367 was detected at HH4 and HH5 during chick development. Relative expression value of miR-367 was normalized to the level of 18s mRNA(G). Bars represent mean±SEM. Student’s t-test: *p<0.05, **p<0.01, and ***p<0.001 vs. HH1.


Reference

References

1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004. 116:281–297.
2. Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet. 2008. 9:102–114.
Article
3. He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004. 5:522–531.
Article
4. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993. 75:843–854.
Article
5. Kozomara A, Griffiths-Jones S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 2011. 39(Database issue):D152–D157.
Article
6. Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T. Identification of tissue-specific microRNAs from mouse. Curr Biol. 2002. 12:735–739.
Article
7. Watanabe T, Takeda A, Mise K, Okuno T, Suzuki T, Minami N, Imai H. Stage-specific expression of microRNAs during Xenopus development. FEBS Lett. 2005. 579:318–324.
8. Marson A, Levine SS, Cole MF, Frampton GM, Brambrink T, Johnstone S, Guenther MG, Johnston WK, Wernig M, Newman J, Calabrese JM, Dennis LM, Volkert TL, Gupta S, Love J, Hannett N, Sharp PA, Bartel DP, Jaenisch R, Young RA. Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell. 2008. 134:521–533.
Article
9. Houbaviy HB, Murray MF, Sharp PA. Embryonic stem cell-specific MicroRNAs. Dev Cell. 2003. 5:351–358.
Article
10. Suh MR, Lee Y, Kim JY, Kim SK, Moon SH, Lee JY, Cha KY, Chung HM, Yoon HS, Moon SY, Kim VN, Kim KS. Human embryonic stem cells express a unique set of microRNAs. Dev Biol. 2004. 270:488–498.
Article
11. Kim KS, Kim JS, Lee MR, Jeong HS, Kim J. A study of microRNAs in silico and in vivo: emerging regulators of embryonic stem cells. FEBS J. 2009. 276:2140–2149.
Article
12. Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y, Zhang Y, Yang W, Gruber PJ, Epstein JA, Morrisey EE. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell. 2011. 8:376–388.
Article
13. Rosa A, Spagnoli FM, Brivanlou AH. The miR-430/427/302 family controls mesendodermal fate specification via species-specific target selection. Dev Cell. 2009. 16:517–527.
Article
14. Wienholds E, Kloosterman WP, Miska E, Alvarez-Saavedra E, Berezikov E, de Bruijn E, Horvitz HR, Kauppinen S, Plasterk RH. MicroRNA expression in zebrafish embryonic development. Science. 2005. 309:310–311.
Article
15. Kloosterman WP, Wienholds E, de Bruijn E, Kauppinen S, Plasterk RH. In situ detection of miRNAs in animal embryos using LNA-modified oligonucleotide probes. Nat Methods. 2006. 3:27–29.
Article
16. Brown WR, Hubbard SJ, Tickle C, Wilson SA. The chicken as a model for large-scale analysis of vertebrate gene function. Nat Rev Genet. 2003. 4:87–98.
Article
17. Hamburger V, Hamilton HL. A series of normal stages in the development of the chick embryo. 1951. Dev Dyn. 1992. 195:231–272.
18. Chen C, Ridzon DA, Broomer AJ, Zhou Z, Lee DH, Nguyen JT, Barbisin M, Xu NL, Mahuvakar VR, Andersen MR, Lao KQ, Livak KJ, Guegler KJ. Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res. 2005. 33:e179.
Article
19. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001. 25:402–408.
Article
20. Rosa A, Brivanlou AH. Regulatory non-coding RNAs in pluripotent stem cells. Int J Mol Sci. 2013. 14:14346–14373.
Article
21. Nieto MA, Sargent MG, Wilkinson DG, Cooke J. Control of cell behavior during vertebrate development by Slug, a zinc finger gene. Science. 1994. 264:835–839.
Article
Full Text Links
  • IJSC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr