Numerous fundamental principles of gene regulation and epigenetics have been unraveled using erythroid cells as a model system. This application will exploit our expertise in erythroid development and chromatin architecture to dissect the diverse activities of CTCF, a central multi-functional transcription factor. CTCF can activate or repress transcription, slow elongation by RNA polymerase 2, form long range chromosomal contacts, block enhancer activity, and form chromatin domain boundaries. CTCF bound sites (CBS) can be tissue-specific or tissue-invariant. A leading question in the field is how does CTCF exert these distinct activities. Other key open questions are how lineage-dependent functions and chromatin binding by CTCF are specified. Utilizing an auxin-inducible CTCF degradation system in an erythroid cell line we found that, unexpectedly, CBS are turned over with highly variable kinetics. This leads to the hypothesis that location- specific differences in susceptibility to degradation may be a reflection of CTCF chromatin residence time that is locally specified by chromatin context. We further hypothesize that such differences in residence times are critical features of distinct CTCF mechanisms of action.
In Specific Aim 1 we will first determine erythroid- and maturation stage-specific CBS genome wide, globally assess auxin-modulated CBS dynamics in both immature and mature erythroid cells, and examine consequences of acute CTCF depletion on gene expression. The determinants of CBS specificity and dynamics will be dissected via genome editing and targeted proteomics (ChIP-SICAP).
In Specific Aim 2, we will test the hypothesis that CBS dynamics are a function of chromatin residence time. This will be measured by single molecule tracking (SMT) of CTCF, to be carried out in collaboration with Dr. Melike Lakadamyali at UPenn. These studies will include perturbative manipulations of CTCF. By combining acute CTCF degradation with PRO-seq we discovered that many genes with CBS near the start site exhibit increased transcription in the anti-sense direction upon CTCF loss. This suggests a novel role of CTCF in governing transcription directionality.
In Specific Aim 3 we will determine the mechanism by which CTCF regulates transcription directionality, exploring chromatin looping versus functioning as an elongation barrier as possible alternative mechanisms. Single molecule RNA FISH will be employed to study the dynamics and mechanisms of transcription directionality. These studies address longstanding questions as to how CTCF accomplishes its diverse functions, including new ones discovered here, and are expected to have broad ramifications for the fields of gene regulation and nuclear architecture.

Public Health Relevance

The formation and differentiation of red blood cells is under the control of many genes. This grant aims to explore how factors controlling gene expression function on a mechanistic basis. A deeper dissection of the gene regulatory circuitry underpinning blood cell formation might aid in a better understanding and treatment of blood diseases.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
2R01DK058044-19
Application #
10050051
Study Section
Molecular and Cellular Hematology Study Section (MCH)
Program Officer
Bishop, Terry Rogers
Project Start
2002-03-01
Project End
2024-04-30
Budget Start
2020-07-15
Budget End
2021-04-30
Support Year
19
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Children's Hospital of Philadelphia
Department
Type
DUNS #
073757627
City
Philadelphia
State
PA
Country
United States
Zip Code
19146
Dogan, Nergiz; Wu, Weisheng; Morrissey, Christapher S et al. (2015) Occupancy by key transcription factors is a more accurate predictor of enhancer activity than histone modifications or chromatin accessibility. Epigenetics Chromatin 8:16
Wu, Weisheng; Morrissey, Christapher S; Keller, Cheryl A et al. (2014) Dynamic shifts in occupancy by TAL1 are guided by GATA factors and drive large-scale reprogramming of gene expression during hematopoiesis. Genome Res 24:1945-62
Pimkin, Maxim; Kossenkov, Andrew V; Mishra, Tejaswini et al. (2014) Divergent functions of hematopoietic transcription factors in lineage priming and differentiation during erythro-megakaryopoiesis. Genome Res 24:1932-44
Deng, Wulan; Blobel, Gerd A (2014) Manipulating nuclear architecture. Curr Opin Genet Dev 25:1-7
Deng, Wulan; Rupon, Jeremy W; Krivega, Ivan et al. (2014) Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell 158:849-860
Lai, Fan; Orom, Ulf A; Cesaroni, Matteo et al. (2013) Activating RNAs associate with Mediator to enhance chromatin architecture and transcription. Nature 494:497-501
Kadauke, Stephan; Udugama, Maheshi I; Pawlicki, Jan M et al. (2012) Tissue-specific mitotic bookmarking by hematopoietic transcription factor GATA1. Cell 150:725-37
Wang, Yuhuan; Meng, Ronghua; Hayes, Vincent et al. (2011) Pleiotropic platelet defects in mice with disrupted FOG1-NuRD interaction. Blood 118:6183-91
Wu, Weisheng; Cheng, Yong; Keller, Cheryl A et al. (2011) Dynamics of the epigenetic landscape during erythroid differentiation after GATA1 restoration. Genome Res 21:1659-71
Lejon, Sara; Thong, Sock Yue; Murthy, Andal et al. (2011) Insights into association of the NuRD complex with FOG-1 from the crystal structure of an RbAp48·FOG-1 complex. J Biol Chem 286:1196-203

Showing the most recent 10 out of 33 publications