In embryonic development, expression of maternal mRNAs regulate cell cycle progression and early developmental processes prior to the onset of zygotic gene transcription. The molecular basis of maternal mRNA translational control is poorly understood. The regulation of protein translation through the cytoplasmic polyadenylation of maternal mRNAs occurs in many organisms including Xenopus, C. elegans, D.melanogaster and the mouse. In Xenopus, progesterone-stimulated cytoplasmic polyadenylation of maternal mRNAs control the synthesis of several key regulators of cell cycle progression (such as Mos, cyclin B1, cyclin B2, cyclin A1 and cdk2). The most extensively characterized of these translationally regulated proteins is the Mos proto-oncogene. Cytoplasmic polyadenylation of Mos mRNA has been shown to be essential for Mos protein synthesis, progesterone-stimulated maturation of Xenopus oocytes and Mos-mediated cytostatic factor activity in mouse oocytes. Despite the recent advances in our knowledge concerning the importance of cytoplasmic polyadenylation in regulation mRNA expression, little is known about the molecular mechanism(s) which mediate this process. The mitogen activated protein kinase (MAPK) signaling pathway (Raf/MEK/MAPK) has been shown to be activated during Xenopus oocyte maturation. Our recent studies have implicated Raf-1 and MEK in the control of Mos protein synthesis through stimulation of the cytoplasmic polyadenylation of Mos mRNA. The studies described in this application will characterize the role of the MAPK signaling pathway in the translational control of Mos and other key regulators of cell cycle regulators of cell cycle progression during vertebrate oocyte maturation. These studies will provide insights into an evolutionary conserved process crucial to early development and yield valuable information on the temporal control of cell cycle progression exerted through the translational regulation of maternal mRNAs. In addition to adding to our understanding of the regulatory pathways governing egg development and fertilization, these studies may provide novel information for the design of therapeutic interventions in the treatment of certain human cancers.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
5R01HD035688-02
Application #
6125580
Study Section
Reproductive Biology Study Section (REB)
Program Officer
Tasca, Richard J
Project Start
1998-12-15
Project End
2001-11-30
Budget Start
1999-12-01
Budget End
2000-11-30
Support Year
2
Fiscal Year
2000
Total Cost
$247,581
Indirect Cost
Name
University of Chicago
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
225410919
City
Chicago
State
IL
Country
United States
Zip Code
60637
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