-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.

3818 PIEZO1-TMEM16F Interplay in Hereditary Xerocytosis

Program: Oral and Poster Abstracts
Session: 101. Red Cells and Erythropoiesis, Excluding Iron: Poster III
Hematology Disease Topics & Pathways:
Research, Fundamental Science, Genetic Disorders, Diseases, Biological Processes, pathogenesis
Monday, December 11, 2023, 6:00 PM-8:00 PM

Pengfei Liang, PhD1, Yang Zhang1*, Yui Chun Serena Wan1*, Shang Ma2*, Ping Dong1*, Augustus Lowry3*, Samuel Francis, MD1, Sanjay Khandelwal, PhD4*, Martha Delahunty, PhD5*, Marilyn J. Telen, MD6, John J Strouse, MD, PhD7, Gowthami Arepally, MD1* and Huanghe Yang, PhD3

1Duke University, Durham
2UT Southwestern Medical Center, Dallas, TX
3Duke University School of Medicine, Durham, NC
4Duke Medical Center, Durham, NC
5Duke Comprehensive Sickle Cell Center, Duke Univ. School of Medicine, Durham, NC
6Duke Univ. Med. Ctr., Durham, NC
7Department of Medicine, Division of Hematology, Duke University School of Medicine, Durham, NC

Cell surface exposure of phosphatidylserine (PS), an anionic phospholipid that is usually confined to the inner leaflet of the plasma membrane, triggers a plethora of cellular responses. PS exposure in RBCs contributes to blood coagulation, promotes RBC aggregation and adhesion to endothelial cells, and accelerates the removal of aging RBCs from circulation. The existence of CaPLSase, a passive phospholipid transporter that catalyzes rapid PS exposure in response to intracellular Ca2+ elevation, was first observed in RBCs in the 1990s. However, the molecular identity of RBC CaPLSase, its activation mechanism and its role in red cell disorders remain elusive. Here we demonstrate that TMEM16F, the long-sought-after RBC CaPLSase, is activated by calcium influx through the mechanosensitive channel PIEZO1 in RBCs. PIEZO1-TMEM16F functional coupling is enhanced in RBCs from individuals with hereditary xerocytosis (HX), a RBC disorder caused by PIEZO1 gain-of-function (GOF) channelopathy. Enhanced PIEZO1-TMEM16F coupling leads to an increased propensity to expose PS, which may contribute to HX clinical manifestations including anemia, splenomegaly and post-splenectomy thrombosis. Pharmacological inhibition of PIEZO1 prevents calcium-induced dehydration, hemolysis and PS exposure in HX RBCs. Our study reveals an activation mechanism of TMEM16F and its pathophysiological function in HX, providing insights into potential treatment.

Disclosures: Telen: CSL Behring: Research Funding. Strouse: Agios, Takeda, Disc Medicine: Consultancy, Research Funding.

*signifies non-member of ASH