1. Academic Validation
  2. Targeting PRMT1-mediated FLT3 methylation disrupts maintenance of MLL-rearranged acute lymphoblastic leukemia

Targeting PRMT1-mediated FLT3 methylation disrupts maintenance of MLL-rearranged acute lymphoblastic leukemia

  • Blood. 2019 Oct 10;134(15):1257-1268. doi: 10.1182/blood.2019002457.
Yinghui Zhu 1 Xin He 1 Yi-Chun Lin 2 Haojie Dong 1 Lei Zhang 3 Xianwei Chen 4 Zhihao Wang 5 Yudao Shen 6 Min Li 7 Hanying Wang 1 Jie Sun 1 Le Xuan Nguyen 1 Han Zhang 2 Wenjuan Jiang 2 Yanzhong Yang 5 Jianjun Chen 8 Markus Müschen 8 Chun-Wei Chen 8 Marina Y Konopleva 9 Weili Sun 10 Jian Jin 6 Nadia Carlesso 1 Guido Marcucci 1 Yun Luo 2 Ling Li 1
Affiliations

Affiliations

  • 1 Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA.
  • 2 College of Pharmacy, Western University of Health Sciences, Pomona, CA.
  • 3 Translational Biomarker Discovery Core.
  • 4 Division of Stem Cell Biology Research, Department of Developmental and Stem Cell Biology, and.
  • 5 Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope Medical Center, Duarte, CA.
  • 6 Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
  • 7 Department of Information Sciences, Beckman Research Institute, City of Hope Medical Center, Duarte, CA.
  • 8 Department of Systems Biology and The Gehr Family Center for Leukemia Research, Beckman Research Institute, City of Hope Medical Center, Monrovia, CA.
  • 9 Department of Leukemia, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX; and.
  • 10 Department of Pediatrics, City of Hope Medical Center, Duarte, CA.
Abstract

Relapse remains the main cause of MLL-rearranged (MLL-r) acute lymphoblastic leukemia (ALL) treatment failure resulting from persistence of drug-resistant clones after conventional chemotherapy treatment or targeted therapy. Thus, defining mechanisms underlying MLL-r ALL maintenance is critical for developing effective therapy. PRMT1, which deposits an asymmetric dimethylarginine mark on histone/non-histone proteins, is reportedly overexpressed in various cancers. Here, we demonstrate elevated PRMT1 levels in MLL-r ALL cells and show that inhibition of PRMT1 significantly suppresses leukemic cell growth and survival. Mechanistically, we reveal that PRMT1 methylates Fms-like receptor tyrosine kinase 3 (FLT3) at arginine (R) residues 972 and 973 (R972/973), and its oncogenic function in MLL-r ALL cells is FLT3 methylation dependent. Both biochemistry and computational analysis demonstrate that R972/973 methylation could facilitate recruitment of adaptor proteins to FLT3 in a phospho-tyrosine (Y) residue 969 (Y969) dependent or independent manner. Cells expressing R972/973 methylation-deficient FLT3 exhibited more robust Apoptosis and growth inhibition than did Y969 phosphorylation-deficient FLT3-transduced cells. We also show that the capacity of the type I PRMT inhibitor MS023 to inhibit leukemia cell viability parallels baseline FLT3 R972/973 methylation levels. Finally, combining FLT3 tyrosine kinase inhibitor PKC412 with MS023 treatment enhanced elimination of MLL-r ALL cells relative to PKC412 treatment alone in patient-derived mouse xenografts. These results indicate that abolishing FLT3 arginine methylation through PRMT1 inhibition represents a promising strategy to target MLL-r ALL cells.

Figures
Products