FBP1-Altered Carbohydrate Metabolism Reduces Leukemic Viability through Activating P53 and Modulating the Mitochondrial Quality Control System In Vitro

Int J Mol Sci. 2022 Sep 27;23(19):11387. doi: 10.3390/ijms231911387.

Abstract

Acute myeloid leukemia (AML)-the most frequent form of adult blood cancer-is characterized by heterogeneous mechanisms and disease progression. Developing an effective therapeutic strategy that targets metabolic homeostasis and energy production in immature leukemic cells (blasts) is essential for overcoming relapse and improving the prognosis of AML patients with different subtypes. With respect to metabolic regulation, fructose-1,6-bisphosphatase 1 (FBP1) is a gluconeogenic enzyme that is vital to carbohydrate metabolism, since gluconeogenesis is the central pathway for the production of important metabolites and energy necessary to maintain normal cellular activities. Beyond its catalytic activity, FBP1 inhibits aerobic glycolysis-known as the "Warburg effect"-in cancer cells. Importantly, while downregulation of FBP1 is associated with carcinogenesis in major human organs, restoration of FBP1 in cancer cells promotes apoptosis and prevents disease progression in solid tumors. Recently, our large-scale sequencing analyses revealed FBP1 as a novel inducible therapeutic target among 17,757 vitamin-D-responsive genes in MV4-11 or MOLM-14 blasts in vitro, both of which were derived from AML patients with FLT3 mutations. To investigate FBP1's anti-leukemic function in this study, we generated a new AML cell line through lentiviral overexpression of an FBP1 transgene in vitro (named FBP1-MV4-11). Results showed that FBP1-MV4-11 blasts are more prone to apoptosis than MV4-11 blasts. Mechanistically, FBP1-MV4-11 blasts have significantly increased gene and protein expression of P53, as confirmed by the P53 promoter assay in vitro. However, enhanced cell death and reduced proliferation of FBP1-MV4-11 blasts could be reversed by supplementation with post-glycolytic metabolites in vitro. Additionally, FBP1-MV4-11 blasts were found to have impaired mitochondrial homeostasis through reduced cytochrome c oxidase subunit 2 (COX2 or MT-CO2) and upregulated PTEN-induced kinase (PINK1) expressions. In summary, this is the first in vitro evidence that FBP1-altered carbohydrate metabolism and FBP1-activated P53 can initiate leukemic death by activating mitochondrial reprogramming in AML blasts, supporting the clinical potential of FBP1-based therapies for AML-like cancers.

Keywords: AML; FBP1; OXPHOS; P53; autophagy; glycolysis; metabolism; mitochondrion; mitophagy.

MeSH terms

  • Apoptosis
  • Carbohydrate Metabolism* / drug effects
  • Carbohydrate Metabolism* / genetics
  • Carbon Dioxide / metabolism
  • Cell Line, Tumor
  • Cyclooxygenase 2 / metabolism
  • Disease Progression
  • Electron Transport Complex IV / metabolism
  • Fructose / pharmacology
  • Fructose-Bisphosphatase / genetics
  • Fructose-Bisphosphatase / metabolism
  • Glycolysis
  • Granulocyte Precursor Cells* / metabolism
  • Humans
  • Leukemia, Myeloid, Acute* / drug therapy
  • Leukemia, Myeloid, Acute* / genetics
  • Leukemia, Myeloid, Acute* / metabolism
  • Mitochondria* / drug effects
  • Mitochondria* / genetics
  • Mitochondria* / metabolism
  • Protein Kinases / metabolism
  • Tumor Suppressor Protein p53* / genetics
  • Tumor Suppressor Protein p53* / metabolism
  • Vitamin D / pharmacology
  • Vitamins / pharmacology
  • fms-Like Tyrosine Kinase 3 / genetics
  • fms-Like Tyrosine Kinase 3 / metabolism

Substances

  • Tumor Suppressor Protein p53
  • Vitamins
  • Vitamin D
  • Carbon Dioxide
  • Fructose
  • Cyclooxygenase 2
  • Electron Transport Complex IV
  • Protein Kinases
  • FLT3 protein, human
  • fms-Like Tyrosine Kinase 3
  • FBP1 protein, human
  • Fructose-Bisphosphatase