Retinogenesis requires a tightly controlled balance of retinal progenitor cell (RPC) proliferation and differentiation. Deregulation of this mechanism often results in profound neuro-developmental disorders or cancer. Our long term goal is to elucidate the transcriptional mechanisms coupling RPC cell cycle regulation to neuronal differentiation. In this proposal, experiments will define the role of the transcriptional regulato Ronin (Thap11) in retinogenesis. Ronin was identified recently as a novel embryonic stem cell (ESC) pluripotency factor, influencing ESC proliferation and growth. Ronin is also expressed throughout the developing brain and retina, but its role in these tissues is unknown. In preliminary studies, we have found that Ronin mutant retinae phenocopy the Cyclin D1 null mice thereby implicating Ronin as a regulator of the cell cycle G1 to S-phase transition. However, analysis of preliminary Ronin retinal ChIP-seq data showed enrichment for mitochondrial genes rather than cell cycle machinery. Recently, mitochondria have emerged as critical regulators of the G1 to S-phase transition in both flies and rodents by promoting Cyclin E activity and entry into S-phase. Therefore, we hypothesize that Ronin influences the RPC cell cycle by directly regulating genes required for proper mitochondrial function and promotion of S-phase entry. To test this hypothesis, we have assembled a team with diverse expertise, to facilitate a synergistic, multi- disciplinary approach using genetic loss- and gain-of-function experiments, metabolic profiling, live retinal microscopy and genomics. Upon completion of this proposal, we expect that we will have identified a new transcriptional mechanism that couples mitochondrial function to cell cycle progression in RPCs. This deeper understanding of the transcriptional regulation of multipotent, proliferative RPCs will ultimately inform efficacious strategies for retinal cell replacement therapies as well as novel cancer drug targets functioning at the interface of mitochondrial bioenergetics and the cell cycle.

Public Health Relevance

Diseases such as retinal degeneration, retinal cancer and blindness are often the result of defects in the regulation of retinal progenitor cell (RPC) proliferation in utero. This proposal aims to elucidate the function of a gene named Ronin (Thap11) which we hypothesize plays a novel and essential role in ensuring that retinal proliferation and differentiation are properly balanced during development. Identification of Ronin as a molecular regulator of the RPC cell cycle may lead to the design of therapeutic interventions to more effectively treat retinal developmental disorders, degeneration and cancer.

Agency
National Institute of Health (NIH)
Institute
National Eye Institute (NEI)
Type
Research Project (R01)
Project #
1R01EY024906-01
Application #
8797939
Study Section
Special Emphasis Panel (BVS)
Program Officer
Neuhold, Lisa
Project Start
2014-12-01
Project End
2019-11-30
Budget Start
2014-12-01
Budget End
2015-11-30
Support Year
1
Fiscal Year
2015
Total Cost
$394,792
Indirect Cost
$144,792
Name
Baylor College of Medicine
Department
Physiology
Type
Schools of Medicine
DUNS #
051113330
City
Houston
State
TX
Country
United States
Zip Code
77030
Barrasso, Anthony P; Wang, Shang; Tong, Xuefei et al. (2018) Live imaging of developing mouse retinal slices. Neural Dev 13:23
Barrasso, Anthony P; Tong, Xuefei; Poché, Ross A (2018) The mito::mKate2 mouse: A far-red fluorescent reporter mouse line for tracking mitochondrial dynamics in vivo. Genesis 56:
Poché, Ross A; Zhang, Min; Rueda, Elda M et al. (2016) RONIN Is an Essential Transcriptional Regulator of Genes Required for Mitochondrial Function in the Developing Retina. Cell Rep 14:1684-1697