학술논문

Nf1 and Sh2b3 mutations cooperate in vivo in a mouse model of juvenile myelomonocytic leukemia
Document Type
article
Source
Blood Advances. 5(18)
Subject
Biomedical and Clinical Sciences
Oncology and Carcinogenesis
Pediatric
Genetics
Pediatric Cancer
Cancer
Pediatric Research Initiative
Hematology
Stem Cell Research
Rare Diseases
Childhood Leukemia
Aetiology
2.1 Biological and endogenous factors
Inflammatory and immune system
Animals
Child
Preschool
Hematopoietic Stem Cell Transplantation
Humans
Leukemia
Myelomonocytic
Juvenile
Mice
Mutation
Prognosis
Signal Transduction
Cardiovascular medicine and haematology
Language
Abstract
Juvenile myelomonocytic leukemia (JMML) is initiated in early childhood by somatic mutations that activate Ras signaling. Although some patients have only a single identifiable oncogenic mutation, others have 1 or more additional alterations. Such secondary mutations, as a group, are associated with an increased risk of relapse after hematopoietic stem cell transplantation or transformation to acute myeloid leukemia. These clinical observations suggest a cooperative effect between initiating and secondary mutations. However, the roles of specific genes in the prognosis or clinical presentation of JMML have not been described. In this study, we investigate the impact of secondary SH2B3 mutations in JMML. We find that patients with SH2B3 mutations have adverse outcomes, as well as higher white blood cell counts and hemoglobin F levels in the peripheral blood. We further demonstrate this interaction in genetically engineered mice. Deletion of Sh2b3 cooperates with conditional Nf1 deletion in a dose-dependent fashion. These studies illustrate that haploinsufficiency for Sh2b3 contributes to the severity of myeloproliferative disease and provide an experimental system for testing treatments for a high-risk cohort of JMML patients.