Your browser doesn't support javascript.
loading
Functional Characterization of Cooperating MGA Mutations in RUNX1::RUNX1T1 Acute Myeloid Leukemia.
Klco, Jeffery; Thomas, Melvin; Qi, Wenqing; Walsh, Michael; Ma, Jing; Westover, Tamara; Abdelhamed, Sherif; Ezzell, Lauren; Rolle, Chandra; Xiong, Emily; Rosikiewicz, Wojciech; Xu, Beisi; Pruett-Miller, Shondra; Loughran, Allister; Janke, Laura.
Afiliação
  • Klco J; St. Jude Children's Research Hospital.
  • Thomas M; St. Jude Children's Research Hospital.
  • Qi W; St. Jude Children's Research Hospital.
  • Walsh M; St. Jude Childrens Hospital.
  • Ma J; St. Jude Children's Research Hospital.
  • Westover T; St. Jude Children's Research Hospital.
  • Abdelhamed S; Seagen.
  • Ezzell L; St. Jude Children's Research Hospital.
  • Rolle C; St. Jude Children's Research Hospital.
  • Xiong E; St. Jude Children's Research Hospital.
  • Rosikiewicz W; St. Jude Children's Research Hospital.
  • Xu B; St Jude Children's Research Hospital.
  • Pruett-Miller S; St. Jude Children's Research Hospital.
  • Loughran A; St. Jude Children's Research Hospital.
  • Janke L; St. Jude Children's Research Hospital.
Res Sq ; 2023 Sep 22.
Article em En | MEDLINE | ID: mdl-37790524
ABSTRACT
MGA (Max-gene associated) is a dual-specificity transcription factor that negatively regulates MYC-target genes to inhibit proliferation and promote differentiation. Loss-of-function mutations in MGA have been commonly identified in several hematological neoplasms, including acute myeloid leukemia (AML) with RUNX1RUNX1T1, however, very little is known about the impact of these MGA alterations on normal hematopoiesis or disease progression. We show that representative MGA mutations identified in patient samples abolish protein-protein interactions and transcriptional activity. Using a series of human and mouse model systems, including a newly developed conditional knock-out mouse strain, we demonstrate that loss of MGA results in upregulation of MYC and E2F targets, cell cycle genes, mTOR signaling, and oxidative phosphorylation in normal hematopoietic cells, leading to enhanced proliferation. The loss of MGA induces an open chromatin state at promotors of genes involved in cell cycle and proliferation. RUNX1RUNX1T1 expression in Mga-deficient murine hematopoietic cells leads to a more aggressive AML with a significantly shortened latency. These data show that MGA regulates multiple pro-proliferative pathways in hematopoietic cells and cooperates with the RUNX1RUNX1 T1 fusion oncoprotein to enhance leukemogenesis.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Ano de publicação: 2023 Tipo de documento: Article