The strict temporal control of maternal mRNA translational activation is critical for vertebrate oocyte maturation and meiotic cell cycle progression. However, the molecular mechanisms regulating the temporal order of mRNA translation have not been established. The frog, Xenopus laevis, has proven to be a useful model system to study the process of mRNA translation. In the frog, the translational induction of the mRNA encoding the Mos proto-oncogene has been shown to occur soon after exposure to the maturation stimulus, progesterone. This early translation of the Mos mRNA is essential for subsequent progression of progesterone-stimulated maturation. We have recently identified an element within the 3' UTR of the Mos mRNA, distinct from the previously described cytoplasmic polyadenylation element (CPE), which mediates this early translational induction. This polyadenylation response element (PRE) is responsive to mitogen-activated protein kinase (MAP kinase) signaling and differs from CPE sequences which direct late mRNA translation in response to maturation promoting factor (MPF) signaling. Our recent data suggest that functional PRE sequences may be present in the 3' UTRs of other Xenopus maternal mRNAs. In this study we will test the hypothesis that PRE-mediated mRNA translational control is the critical regulatory step required for the progesterone-triggered all-or-none cell fate transition from an immature to a mature oocyte. We will characterize the molecular basis for PRE-mediated translational activation and elucidate the combinatorial effects of the PRE and CPE regulatory processes on the temporal control of maternal mRNA translation. Lastly, we will characterize the signal transduction pathways which mediate induction of PRE-dependent mRNA translation in response to the initial, Mos-independent, progesterone stimulus. These studies will further our understanding of the coordinated action of distinct signaling pathways in the temporal control of maternal mRNA translation. ? ?

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
5R01HD035688-07
Application #
6898686
Study Section
Reproductive Biology Study Section (REB)
Program Officer
Tasca, Richard J
Project Start
2003-07-01
Project End
2007-07-14
Budget Start
2005-07-01
Budget End
2007-07-14
Support Year
7
Fiscal Year
2005
Total Cost
$222,557
Indirect Cost
Name
University of Arkansas for Medical Sciences
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
122452563
City
Little Rock
State
AR
Country
United States
Zip Code
72205
MacNicol, Melanie C; Cragle, Chad E; McDaniel, F Kennedy et al. (2017) Evasion of regulatory phosphorylation by an alternatively spliced isoform of Musashi2. Sci Rep 7:11503
MacNicol, Melanie C; Cragle, Chad E; Arumugam, Karthik et al. (2015) Functional Integration of mRNA Translational Control Programs. Biomolecules 5:1580-99
MacNicol, Angus M; Hardy, Linda L; Spencer, Horace J et al. (2015) Neural stem and progenitor cell fate transition requires regulation of Musashi1 function. BMC Dev Biol 15:15
Janganati, Venumadhav; Penthala, Narsimha Reddy; Cragle, Chad E et al. (2014) Heterocyclic aminoparthenolide derivatives modulate G(2)-M cell cycle progression during Xenopus oocyte maturation. Bioorg Med Chem Lett 24:1963-7
Cragle, Chad; MacNicol, Angus M (2014) Musashi protein-directed translational activation of target mRNAs is mediated by the poly(A) polymerase, germ line development defective-2. J Biol Chem 289:14239-51
Rutledge, Charlotte E; Lau, Ho-Tak; Mangan, Hazel et al. (2014) Efficient translation of Dnmt1 requires cytoplasmic polyadenylation and Musashi binding elements. PLoS One 9:e88385
Penthala, Narsimha R; Bommagani, Shobanbabu; Janganati, Venumadhav et al. (2014) Heck products of parthenolide and melampomagnolide-B as anticancer modulators that modify cell cycle progression. Eur J Med Chem 85:517-25
Arumugam, Karthik; MacNicol, Melanie C; Wang, Yiying et al. (2012) Ringo/cyclin-dependent kinase and mitogen-activated protein kinase signaling pathways regulate the activity of the cell fate determinant Musashi to promote cell cycle re-entry in Xenopus oocytes. J Biol Chem 287:10639-49
Charlesworth, Amanda; Yamamoto, Tomomi M; Cook, Jonathan M et al. (2012) Xenopus laevis zygote arrest 2 (zar2) encodes a zinc finger RNA-binding protein that binds to the translational control sequence in the maternal Wee1 mRNA and regulates translation. Dev Biol 369:177-90
Arumugam, Karthik; Macnicol, Melanie C; Macnicol, Angus M (2012) Autoregulation of Musashi1 mRNA translation during Xenopus oocyte maturation. Mol Reprod Dev 79:553-63

Showing the most recent 10 out of 22 publications