-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

3590 Thyroid Hormone Receptor Beta and NCOA4 Regulate Terminal Erythrocyte Differentiation

Hematopoiesis: Cytokines, Signal Transduction, Apoptosis and Cell Cycle Regulation
Program: Oral and Poster Abstracts
Session: 504. Hematopoiesis: Cytokines, Signal Transduction, Apoptosis and Cell Cycle Regulation: Poster III
Monday, December 7, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Hsiang-Ying Lee, Ph.D.1*, Harvey F. Lodish, Ph.D.2, Xiaofei Gao1*, Wenbo Li3*, Randy Platt4*, Inmaculada Barrasa1*, Russell Elmes1* and Michael G. Rosenfeld3*

1Whitehead Institute for Biomedical Research, Cambridge
2Massachusetts Institute of Technology, Whitehead Institute for Biomedical Research, Cambridge, MA
3UCSD, San Diego
4Broad Institute, Cambridge

An effect of thyroid hormone on erythropoiesis has been known for more than a century but the molecular mechanism(s) by which thyroid hormone affects red cell formation are still elusive. Our study demonstrates an essential role of thyroid hormone during terminal human erythroid cell differentiation; specific depletion of thyroid hormone from the culture medium completely blocked human erythroid differentiation. Genome wide analysis showed that thyroid hormone receptor β (TRβ) occupies many gene loci encoding transcripts abundant during terminal erythropoiesis, including globin genes, and cooperates with GATA-1 and RNA polymerase II (Pol II) to regulate their expression. Treatment with TRβ agonists enhanced erythroblast differentiation in vivo and alleviated anemic symptoms in a chronic anemia mouse model. To identify factors that cooperate with TRβ during human erythroid terminal differentiation, we conducted RNA-Seq in human reticulocytes and identified NCOA4 as a critical regulator of terminal differentiation. Furthermore, Ncoa4-/- mice are anemic in both the embryonic and perinatal periods and fail to respond to thyroid hormone by enhanced erythropoiesis. Genome wide analysis suggests that thyroid hormone promotes NCOA4 recruitment to chromatin regions that are in proximity to Pol II and are highly associated with transcripts abundant during terminal differentiation. Collectively, our results reveal the molecular mechanism of thyroid hormone function on red blood cell formation and are potentially useful to treat certain anemias.

Disclosures: No relevant conflicts of interest to declare.

*signifies non-member of ASH