Abstract

Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative neoplasm in children characterized by the overproduction of monocytic cells that infiltrate the spleen, lung, and liver. JMML remains a disease for which few curative therapies are available other than myeloablative hematopoietic stem cell transplant (HSCT); however, relapse remains a major cause of treatment failure and the long-term morbidities of HSCT for survivors are substantial. A hallmark feature of JMML is acquired hypersensitivity by clonal myeloid progenitor cells to granulocyte macrophage-colony stimulating factor (GM-CSF) via a largely unknown mechanism. Here, we identify c-Cbl (henceforth referred to as Cbl) as a GM-CSF receptor (GMR) adaptor protein that targets Src for ubiquitin-mediated destruction upon GM-CSF stimulation and show that a loss of negative regulation of Src is pivotal in the hyperactivation of GMR signaling in Cbl-mutated JMML cells. Notably, dasatinib, an U.S. Food and Drug Administration-approved multikinase inhibitor that also targets Src family, dramatically attenuated the spontaneous and GM-CSF-induced hypersensitive growth phenotype of mononuclear cells from peripheral blood and bone marrow collected from JMML patients harboring Cbl or other known JMML-associated mutations. These findings reveal Src kinase as a critical oncogenic driver underlying JMML.

Highlights

  • The majority of juvenile myelomonocytic leukemia (JMML)– associated mutations have been mapped to genes that encode proteins that signal through the Ras/mitogen-activated protein kinase (MAPK) pathway, including NF1, NRAS, KRAS, and PTPN11 (SHP2), which account for approximately 75% of JMML patients [1]

  • GM-CSF receptor (GMR) is composed of a ligand-specific a chain (GMRa) and the b common (GMRbc) signaling subunit, which is shared with the IL-3 and IL-5 receptors [21]

  • Mutations in Cbl were predicted to result in the loss of E3 function [3, 7], the molecular mechanism by which these mutations cause JMML and more precisely the observed granulocyte macrophage-colony stimulating factor (GM-CSF) hypersensitivity that underlies JMML remained unclear

Read more

Summary

Introduction

The majority of juvenile myelomonocytic leukemia (JMML)– associated mutations have been mapped to genes that encode proteins that signal through the Ras/mitogen-activated protein kinase (MAPK) pathway, including NF1, NRAS, KRAS, and PTPN11 (SHP2), which account for approximately 75% of JMML patients [1]. Cbl contains a highly conserved N-terminal tyrosine kinase-binding (TKB) domain that mediates interaction between Cbl and phosphorylated tyrosines on activated receptor tyrosine kinases such as epidermal growth factor receptor Y371 within the linker region becomes phosphorylated upon interaction with tyrosine kinases such as EGFR and insulin receptor, leading to conformational change in Cbl and activation of its E3 activity [13]. Cbl's E3 function serves as a negative regulator of signaling, its role as an adaptor molecule potentiates signal

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.