The coordination of cell division with differentiation is critical for appropriate retinal formation. If this process is disrupted, vision may be compromised, and in some cases, malignant retinoblastoma may form. The Rb family of proteins (Rb, p107, and p130) lies at the heart of the regulatory network that controls proliferation during development and the suppression of tumorigenesis. Based on our preliminary data, we hypothesize that these proteins play distinct roles regulating proliferation and differentiation during mouse retinal development. This hypothesis stands in contrast to the long-standing assumption that Rb and p107 play redundant roles in the murine retina. In humans, we suggest that the complete absence of any role for p107 and a minimal role for p130 in regulating cell cycle exit during retinal development results in a unique susceptibility to early childhood retinal tumors.
In Specific Aim 1, we will study the roles played by each Rb family member in regulating retinal progenitor cell proliferation using mutant mice, recombinant retroviral vectors and retinal explant cultures specifically optimized for such studies. Particular attention will be directed toward elucidating redundant vs. compensatory mechanisms among family members.
In Specific Aim 2, gain- and loss-of-function studies will be performed to determine the specific requirement of each Rb family member in the specification and differentiation of the seven classes of retinal cell types. Emphasis will be placed on distinguishing changes in retinal development due to proliferation alterations from direct effects on developmental processes.
In Specific Aim 3, we will compare our data on retinal development in the mouse to developmental data obtained from normal human and monkey samples, along with human retinoblastoma samples to address the question of human susceptibility to retinal tumors. Specific mechanisms underlying retinoblastoma formation will be tested in experiments designed to faithfully model retinoblastoma in the mouse. These studies bring together an extensive knowledge base and reagents from the fields of cell cycle control and retinal development to address fundamental questions regarding retinal development and disease.

Agency
National Institute of Health (NIH)
Institute
National Eye Institute (NEI)
Type
Research Project (R01)
Project #
5R01EY014867-05
Application #
7287391
Study Section
Special Emphasis Panel (ZRG1-VISC (01))
Program Officer
Mariani, Andrew P
Project Start
2003-09-30
Project End
2008-06-30
Budget Start
2007-07-01
Budget End
2008-06-30
Support Year
5
Fiscal Year
2007
Total Cost
$394,725
Indirect Cost
Name
St. Jude Children's Research Hospital
Department
Type
DUNS #
067717892
City
Memphis
State
TN
Country
United States
Zip Code
38105
Stewart, Elizabeth; McEvoy, Justina; Wang, Hong et al. (2018) Identification of Therapeutic Targets in Rhabdomyosarcoma through Integrated Genomic, Epigenomic, and Proteomic Analyses. Cancer Cell 34:411-426.e19
Wang, Lu; Hiler, Daniel; Xu, Beisi et al. (2018) Retinal Cell Type DNA Methylation and Histone Modifications Predict Reprogramming Efficiency and Retinogenesis in 3D Organoid Cultures. Cell Rep 22:2601-2614
Aldiri, Issam; Xu, Beisi; Wang, Lu et al. (2017) The Dynamic Epigenetic Landscape of the Retina During Development, Reprogramming, and Tumorigenesis. Neuron 94:550-568.e10
Stewart, Elizabeth; Federico, Sara M; Chen, Xiang et al. (2017) Orthotopic patient-derived xenografts of paediatric solid tumours. Nature 549:96-100
Hiler, Daniel J; Barabas, Marie E; Griffiths, Lyra M et al. (2016) Reprogramming of mouse retinal neurons and standardized quantification of their differentiation in 3D retinal cultures. Nat Protoc 11:1955-1976
Stewart, Elizabeth; Federico, Sara; Karlstrom, Asa et al. (2016) The Childhood Solid Tumor Network: A new resource for the developmental biology and oncology research communities. Dev Biol 411:287-293
Valle-GarcĂ­a, David; Qadeer, Zulekha A; McHugh, Domhnall S et al. (2016) ATRX binds to atypical chromatin domains at the 3' exons of zinc finger genes to preserve H3K9me3 enrichment. Epigenetics 11:398-414
Dyer, Michael A (2016) Lessons from Retinoblastoma: Implications for Cancer, Development, Evolution, and Regenerative Medicine. Trends Mol Med 22:863-876
Chen, X; Pappo, A; Dyer, M A (2015) Pediatric solid tumor genomics and developmental pliancy. Oncogene 34:5207-15
Stewart, Elizabeth; Shelat, Anang; Bradley, Cori et al. (2015) Development and characterization of a human orthotopic neuroblastoma xenograft. Dev Biol 407:344-55

Showing the most recent 10 out of 41 publications